Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(12): 105378, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37866635

RESUMO

Membrane contact sites (MCSs) between endosomes and the endoplasmic reticulum (ER) are thought to act as specialized trigger zones for Ca2+ signaling, where local Ca2+ released via endolysosomal ion channels is amplified by ER Ca2+-sensitive Ca2+ channels into global Ca2+ signals. Such amplification is integral to the action of the second messenger, nicotinic acid adenine dinucleotide phosphate (NAADP). However, functional regulators of inter-organellar Ca2+ crosstalk between endosomes and the ER remain poorly defined. Here, we identify progesterone receptor membrane component 1 (PGRMC1), an ER transmembrane protein that undergoes a unique heme-dependent dimerization, as an interactor of the endosomal two pore channel, TPC1. NAADP-dependent Ca2+ signals were potentiated by PGRMC1 overexpression through enhanced functional coupling between endosomal and ER Ca2+ stores and inhibited upon PGRMC1 knockdown. Point mutants in PGMRC1 or pharmacological manipulations that reduced its interaction with TPC1 were without effect. PGRMC1 therefore serves as a TPC1 interactor that regulates ER-endosomal coupling with functional implications for cellular Ca2+ dynamics and potentially the distribution of heme.


Assuntos
Sinalização do Cálcio , Retículo Endoplasmático , Endossomos , Receptores de Progesterona , Humanos , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Heme/metabolismo , Lisossomos/metabolismo , NADP/metabolismo , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
2.
J Cell Sci ; 128(2): 232-8, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416817

RESUMO

Two-pore channels (TPCs) are endolysosomal ion channels implicated in Ca(2+) signalling from acidic organelles. The relevance of these ubiquitous proteins for human disease, however, is unclear. Here, we report that lysosomes are enlarged and aggregated in fibroblasts from Parkinson disease patients with the common G2019S mutation in LRRK2. Defects were corrected by molecular silencing of TPC2, pharmacological inhibition of TPC regulators [Rab7, NAADP and PtdIns(3,5)P2] and buffering local Ca(2+) increases. NAADP-evoked Ca(2+) signals were exaggerated in diseased cells. TPC2 is thus a potential drug target within a pathogenic LRRK2 cascade that disrupts Ca(2+)-dependent trafficking in Parkinson disease.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/genética , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/metabolismo , Cálcio/metabolismo , Canais de Cálcio/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Células HEK293 , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Lisossomos/metabolismo , Lisossomos/patologia , NADP/análogos & derivados , NADP/genética , NADP/metabolismo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/genética
3.
Proc Natl Acad Sci U S A ; 111(36): 13087-92, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25157141

RESUMO

The two-pore channels (TPC1 and TPC2) belong to an ancient family of intracellular ion channels expressed in the endolysosomal system. Little is known about how regulatory inputs converge to modulate TPC activity, and proposed activation mechanisms are controversial. Here, we compiled a proteomic characterization of the human TPC interactome, which revealed that TPCs complex with many proteins involved in Ca(2+) homeostasis, trafficking, and membrane organization. Among these interactors, TPCs were resolved to scaffold Rab GTPases and regulate endomembrane dynamics in an isoform-specific manner. TPC2, but not TPC1, caused a proliferation of endolysosomal structures, dysregulating intracellular trafficking, and cellular pigmentation. These outcomes required both TPC2 and Rab activity, as well as their interactivity, because TPC2 mutants that were inactive, or rerouted away from their endogenous expression locale, or deficient in Rab binding, failed to replicate these outcomes. Nicotinic acid adenine dinucleotide phosphate (NAADP)-evoked Ca(2+) release was also impaired using either a Rab binding-defective TPC2 mutant or a Rab inhibitor. These data suggest a fundamental role for the ancient TPC complex in trafficking that holds relevance for lysosomal proliferative scenarios observed in disease.


Assuntos
Canais de Cálcio/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Pigmentação , Animais , Sinalização do Cálcio , Proliferação de Células , Cromatografia de Afinidade , Células HEK293 , Humanos , NADP/análogos & derivados , NADP/metabolismo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Reprodutibilidade dos Testes , Xenopus , Proteínas rab de Ligação ao GTP/metabolismo
4.
J Biol Chem ; 288(1): 355-67, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23148228

RESUMO

Changes in cytoplasmic Ca(2+) concentration, resulting from activation of intracellular Ca(2+) channels within the endoplasmic reticulum, regulate several aspects of cellular growth and differentiation. Ca(2+) homeostasis endoplasmic reticulum protein (CHERP) is a ubiquitously expressed protein that has been proposed as a regulator of both major families of endoplasmic reticulum Ca(2+) channels, inositol 1,4,5-trisphosphate receptors (IP(3)Rs) and ryanodine receptors (RyRs), with resulting effects on mitotic cycling. However, the manner by which CHERP regulates intracellular Ca(2+) channels to impact cellular growth is unknown. Here, we challenge previous findings that CHERP acts as a direct cytoplasmic regulator of IP(3)Rs and RyRs and propose that CHERP acts in the nucleus to impact cellular proliferation by regulating the function of the U2 snRNA spliceosomal complex. The previously reported effects of CHERP on cellular growth therefore are likely indirect effects of altered spliceosomal function, consistent with prior data showing that loss of function of U2 snRNP components can interfere with cell growth and induce cell cycle arrest.


Assuntos
Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Proteínas de Membrana/metabolismo , Nucleossomos/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Sequência de Aminoácidos , Sinalização do Cálcio , Ciclo Celular , Membrana Celular/metabolismo , Citoplasma/metabolismo , Retículo Endoplasmático/metabolismo , Células HEK293 , Homeostase , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Jurkat , Dados de Sequência Molecular , Mutação , Interferência de RNA , Ribonucleoproteínas/química , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Spliceossomos/metabolismo , Frações Subcelulares/metabolismo
5.
J Biol Chem ; 287(4): 2296-307, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22117075

RESUMO

Nicotinic acid adenine dinucleotide phosphate (NAADP) is an agonist-generated second messenger that releases Ca(2+) from intracellular acidic Ca(2+) stores. Recent evidence has identified the two-pore channels (TPCs) within the endolysosomal system as NAADP-regulated Ca(2+) channels that release organellar Ca(2+) in response to NAADP. However, little is known about the mechanism coupling NAADP binding to calcium release. To identify the NAADP binding site, we employed a photoaffinity labeling method using a radioactive photoprobe based on 5-azido-NAADP ([(32)P-5N(3)]NAADP) that exhibits high affinity binding to NAADP receptors. In several systems that are widely used for studying NAADP-evoked Ca(2+) signaling, including sea urchin eggs, human cell lines (HEK293, SKBR3), and mouse pancreas, 5N(3)-NAADP selectively labeled low molecular weight sites that exhibited the diagnostic pharmacology of NAADP-sensitive Ca(2+) release. Surprisingly, we were unable to demonstrate labeling of endogenous, or overexpressed, TPCs. Furthermore, labeling of high affinity NAADP binding sites was preserved in pancreatic samples from TPC1 and TPC2 knock-out mice. These photolabeling data suggest that an accessory component within a larger TPC complex is responsible for binding NAADP that is unique from the core channel itself. This observation necessitates critical evaluation of current models of NAADP-triggered activation of the TPC family.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Ativação do Canal Iônico/fisiologia , NADP/análogos & derivados , Pâncreas/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , NADP/metabolismo , Marcadores de Fotoafinidade/química
6.
J Biol Chem ; 287(4): 2308-15, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22117077

RESUMO

Nicotinic acid adenine dinucleotide phosphate (NAADP) is a messenger that regulates calcium release from intracellular acidic stores. Recent studies have identified two-pore channels (TPCs) as endolysosomal channels that are regulated by NAADP; however, the nature of the NAADP receptor binding site is unknown. To further study NAADP binding sites, we have synthesized and characterized [(32)P-5-azido]nicotinic acid adenine dinucleotide phosphate ([(32)P-5N(3)]NAADP) as a photoaffinity probe. Photolysis of sea urchin egg homogenates preincubated with [(32)P-5N(3)]NAADP resulted in specific labeling of 45-, 40-, and 30-kDa proteins, which was prevented by inclusion of nanomolar concentrations of unlabeled NAADP or 5N(3)-NAADP, but not by micromolar concentrations of structurally related nucleotides such as NAD, nicotinic acid adenine dinucleotide, nicotinamide mononucleotide, nicotinic acid, or nicotinamide. [(32)P-5N(3)]NAADP binding was saturable and displayed high affinity (K(d) ∼10 nM) in both binding and photolabeling experiments. [(32)P-5N(3)]NAADP photolabeling was irreversible in a high K(+) buffer, a hallmark feature of NAADP binding in the egg system. The proteins photolabeled by [(32)P-5N(3)]NAADP have molecular masses smaller than the sea urchin TPCs, and antibodies to TPCs do not detect any immunoreactivity that comigrates with either the 45-kDa or the 40-kDa photolabeled proteins. Interestingly, antibodies to TPC1 and TPC3 were able to immunoprecipitate a small fraction of the 45- and 40-kDa photolabeled proteins, suggesting that these proteins associate with TPCs. These data suggest that high affinity NAADP binding sites are distinct from TPCs.


Assuntos
Canais de Cálcio/metabolismo , NADP/análogos & derivados , Óvulo/metabolismo , Strongylocentrotus purpuratus/metabolismo , Animais , Sítios de Ligação , NADP/metabolismo , Marcadores de Fotoafinidade/química , Ligação Proteica
7.
Int J Biol Macromol ; 230: 123205, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36632962

RESUMO

The human sodium-dependent vitamin C transporter-1 (hSVCT1) is localized at the apical membrane domain of polarized intestinal and renal epithelial cells to mediate ascorbic acid (AA) uptake. Currently, little is known about the array of interacting proteins that aid hSVCT1 trafficking and functional expression at the cell surface. Here we used an affinity tagging ('One-STrEP') and proteomic approach to identify hSVCT1 interacting proteins, which resolved secretory carrier-associated membrane protein-2 (SCAMP2) as a novel accessary protein partner. SCAMP2 was validated as an accessory protein by co-immunoprecipitation with hSVCT1. Co-expression of hSVCT1 and SCAMP2 in HEK-293 cells revealed both proteins co-localized in intracellular structures and at the plasma membrane. Functionally, over-expression of SCAMP2 potentiated 14C-AA uptake, and reciprocally silencing endogenous SCAMP2 decreased 14C-AA uptake. Finally, knockdown of endogenous hSVCT1 or SCAMP2 impaired differentiation of human-induced pluripotent stem cells (hiPSCs) toward a neuronal fate. These results establish SCAMP2 as a novel hSVCT1 accessary protein partner that regulates AA uptake in absorptive epithelia and during neurogenesis.


Assuntos
Proteômica , Transportadores de Sódio Acoplados à Vitamina C , Humanos , Células HEK293 , Membrana Celular/metabolismo , Transportadores de Sódio Acoplados à Vitamina C/genética , Transportadores de Sódio Acoplados à Vitamina C/metabolismo , Ácido Ascórbico/farmacologia , Ácido Ascórbico/metabolismo , Neurônios/metabolismo , Transporte Proteico , Proteínas de Transporte/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
8.
PLoS One ; 12(10): e0186541, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29020070

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0073503.].

9.
Cold Spring Harb Protoc ; 2013(3)2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23457336

RESUMO

In the four decades since the Xenopus oocyte was first demonstrated to have the capacity to translate exogenous mRNAs, this system has been exploited for many different experimental purposes. Typically, the oocyte is used either as a "biological test tube" for heterologous expression of proteins without any particular cell biological insight or, alternatively, it is used for applications where cell biology is paramount, such as investigations of the cellular adaptations that power early development. In this article, we discuss the utility of the Xenopus oocyte for studying Ca(2+) signaling in both these contexts.


Assuntos
Cálcio/metabolismo , Técnicas Citológicas/métodos , Oócitos/fisiologia , Transdução de Sinais , Xenopus/fisiologia , Animais
10.
Cold Spring Harb Protoc ; 2013(3)2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23457340

RESUMO

The Xenopus oocyte is frequently used for heterologous expression and for studying the spatiotemporal patterning of Ca(2+) signals. Here, we outline a protocol for nuclear microinjection of the Xenopus oocyte for the purpose of studying how subsequently expressed proteins impact intracellular Ca(2+) signals evoked by inositol trisphosphate (InsP3). Injected oocytes can easily be identified by reporter technologies and the impact of heterologously expressed proteins on the generation and properties of InsP3-evoked Ca(2+) signals can be resolved using caged InsP3 and fluorescent Ca(2+) indicators.


Assuntos
Cálcio/metabolismo , Microinjeções/métodos , Biologia Molecular/métodos , Oócitos/metabolismo , Transdução de Sinais , Animais , Fosfatos de Inositol/metabolismo , Proteínas Recombinantes/biossíntese , Xenopus
11.
Cold Spring Harb Protoc ; 2013(3)2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23457341

RESUMO

Often experimentalists require a quantitative assessment of the levels of heterologously expressed proteins to best interpret changed Ca(2+) signaling patterns. Here, we detail a rapid and convenient western blotting method for individual Xenopus oocytes. The method exploits recently introduced rapid blotting systems, commercially available from Invitrogen (iBlot) or Bio-Rad (Trans-Blot Turbo). The key advantage is speed: from live cell to transferred membrane in <1 h. Therefore, oocytes can be conveniently processed for western blotting to assess relative expression levels, even after a long day of Ca(2+) imaging experiments.


Assuntos
Western Blotting/métodos , Biologia Molecular/métodos , Oócitos/química , Proteínas Recombinantes/análise , Animais , Xenopus
12.
PLoS One ; 8(8): e73503, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24023687

RESUMO

Mammalian cells obtain vitamin B1 (thiamin) from their surrounding environment and convert it to thiamin pyrophosphate (TPP) in the cytoplasm. Most of TPP is then transported into the mitochondria via a carrier-mediated process that involves the mitochondrial thiamin pyrophosphate transporter (MTPPT). Knowledge about the physiological parameters of the MTPP-mediated uptake process, MTPPT targeting and the impact of clinical mutations in MTPPT in patients with Amish lethal microcephaly and neuropathy and bilateral striatal necrosis are not fully elucidated, and thus, were addressed in this study using custom-made (3)H-TPP as a substrate and mitochondria isolated from mouse liver and human-derived liver HepG2 cells. Results showed (3)H-TPP uptake by mouse liver mitochondria to be pH-independent, saturable (Km = 6.79±0.53 µM), and specific for TPP. MTPPT protein was expressed in mouse liver and HepG2 cells, and confocal images showed a human (h)MTPPT-GFP construct to be targeted to mitochondria of HepG2 cells. A serial truncation analysis revealed that all three modules of hMTPPT protein cooperated (although at different levels of efficiency) in mitochondrial targeting rather than acting autonomously as independent targeting module. Finally, the hMTPPT clinical mutants (G125S and G177A) showed proper mitochondrial targeting but displayed significant inhibition in (3)H-TPP uptake and a decrease in level of expression of the MTPPT protein. These findings advance our knowledge of the physiology and cell biology of the mitochondrial TPP uptake process. The results also show that clinical mutations in the hMTPPT system impair its functionality via affecting its level of expression with no effect on its targeting to mitochondria.


Assuntos
Mitocôndrias Hepáticas/metabolismo , Tiamina Pirofosfato/metabolismo , Animais , Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Feminino , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células Hep G2 , Humanos , Cinética , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Proteínas de Transporte da Membrana Mitocondrial , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Proteínas Mutantes/metabolismo , Mutação/genética , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Trítio/metabolismo , Xenopus laevis
13.
Messenger (Los Angel) ; 1(1): 63-76, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25309835

RESUMO

NAADP is a potent Ca2+ mobilizing messenger in a variety of cells but its molecular mechanism of action is incompletely understood. Accumulating evidence indicates that the poorly characterized two-pore channels (TPCs) in animals are NAADP sensitive Ca2+-permeable channels. TPCs localize to the endo-lysosomal system but are functionally coupled to the better characterized endoplasmic reticulum Ca2+ channels to generate physiologically relevant complex Ca2+ signals. Whether TPCs directly bind NAADP is not clear. Here we discuss the idea based on recent studies that TPCs are the pore-forming subunits of a protein complex that includes tightly associated, low molecular weight NAADP-binding proteins.

14.
Messenger (Los Angel) ; 1(1): 86-94, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24829846

RESUMO

Nicotinic acid adenine dinucleotide phosphate (NAADP) is a messenger that regulates calcium release from intracellular acidic stores. Although several channels, including two-pore channels (TPC), ryanodine receptor (RYR) and mucolipin (TRP-ML1) have been implicated in NAADP regulation of calcium signaling, the NAADP receptor has not been identified. In this study, the photoaffinity probe, [32P]-5-azido-NAADP ([32P]-5-N3-NAADP), was used to study NAADP binding proteins in extracts from NAADP responsive Jurkat T-lymphocytes. [32P]-5-N3-NAADP photolabeling of Jurkat S100 cytosolic fractions resulted in the labeling of at least ten distinct proteins. Several of these S100 proteins, including a doublet at 22/23 kDa and small protein at 15 kDa displayed selectivity for NAADP as the labeling was protected by inclusion of unlabeled NAADP, whereas the structurally similar NADP required much higher concentrations for protection. Interestingly, the labeling of several S100 proteins (60, 45, 33 and 28 kDa) was stimulated by low concentrations of unlabeled NAADP, but not by NADP. The effect of NAADP on the labeling of the 60 kDa protein was biphasic, peaking at 100 nM with a five-fold increase and displaying no change at 1 µM NAADP. Several proteins were also photolabeled when the P100 membrane fraction from Jurkat cells was examined. Similar to the results with S100, a 22/23 kDa doublet and a 15 kDa protein appeared to be selectively labeled. NAADP did not increase the labeling of any P100 proteins as it did in the S100 fraction. The photolabeled S100 and P100 proteins were successfully resolved by two-dimensional gel electrophoresis. [32P]-5-N3-NAADP photolabeling and two-dimensional electrophoresis should represent a suitable strategy in which to identify and characterize NAADP binding proteins.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA