Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Cell ; 81(2): 398-407.e4, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33340489

RESUMO

Mechanistic target of rapamycin complex 1 (mTORC1) controls cell growth and proliferation by sensing fluctuations in environmental cues such as nutrients, growth factors, and energy levels. The Rag GTPases (Rags) serve as a critical module that signals amino acid (AA) availability to modulate mTORC1 localization and activity. Recent studies have demonstrated how AAs regulate mTORC1 activity through Rags. Here, we uncover an unconventional pathway that activates mTORC1 in response to variations in threonine (Thr) levels via mitochondrial threonyl-tRNA synthetase TARS2. TARS2 interacts with inactive Rags, particularly GTP-RagC, leading to increased GTP loading of RagA. mTORC1 activity in cells lacking TARS2 is resistant to Thr repletion, showing that TARS2 is necessary for Thr-dependent mTORC1 activation. The requirement of TARS2, but not cytoplasmic threonyl-tRNA synthetase TARS, for this effect demonstrates an additional layer of complexity in the regulation of mTORC1 activity.


Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Mitocôndrias/metabolismo , Proteínas Monoméricas de Ligação ao GTP/genética , Treonina-tRNA Ligase/genética , Treonina/metabolismo , Regulação da Expressão Gênica , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Regulatória Associada a mTOR/genética , Proteína Regulatória Associada a mTOR/metabolismo , Transdução de Sinais , Treonina-tRNA Ligase/antagonistas & inibidores , Treonina-tRNA Ligase/metabolismo
2.
Nature ; 595(7865): 120-124, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34079125

RESUMO

Compartmentalization is a defining characteristic of eukaryotic cells, and partitions distinct biochemical processes into discrete subcellular locations. Microscopy1 and biochemical fractionation coupled with mass spectrometry2-4 have defined the proteomes of a variety of different organelles, but many intracellular compartments have remained refractory to such approaches. Proximity-dependent biotinylation techniques such as BioID provide an alternative approach to define the composition of cellular compartments in living cells5-7. Here we present a BioID-based map of a human cell on the basis of 192 subcellular markers, and define the intracellular locations of 4,145 unique proteins in HEK293 cells. Our localization predictions exceed the specificity of previous approaches, and enabled the discovery of proteins at the interface between the mitochondrial outer membrane and the endoplasmic reticulum that are crucial for mitochondrial homeostasis. On the basis of this dataset, we created humancellmap.org as a community resource that provides online tools for localization analysis of user BioID data, and demonstrate how this resource can be used to understand BioID results better.


Assuntos
Biotinilação , Compartimento Celular , Transporte Proteico , Proteoma/análise , Proteoma/química , Células Cultivadas , Conjuntos de Dados como Assunto , Retículo Endoplasmático/química , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Homeostase , Humanos , Espectrometria de Massas , Mitocôndrias/química , Mitocôndrias/metabolismo , Organelas/química , Organelas/metabolismo , Proteoma/metabolismo , Reprodutibilidade dos Testes
3.
J Biol Chem ; 299(12): 105416, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37918808

RESUMO

Proteostasis requires oxidative metabolism (ATP) and mitigation of the associated damage by glutathione, in an increasingly dysfunctional relationship with aging. SLC3A2 (4F2hc, CD98) plays a role as a disulfide-linked adaptor to the SLC7A5 and SLC7A11 exchangers which import essential amino acids and cystine while exporting Gln and Glu, respectively. The positions of N-glycosylation sites on SLC3A2 have evolved with the emergence of primates, presumably in synchrony with metabolism. Herein, we report that each of the four sites in SLC3A2 has distinct profiles of Golgi-modified N-glycans. N-glycans at the primate-derived site N381 stabilized SLC3A2 in the galectin-3 lattice against coated-pit endocytosis, while N365, the site nearest the membrane promoted glycolipid-galectin-3 (GL-Lect)-driven endocytosis. Our results indicate that surface retention and endocytosis are precisely balanced by the number, position, and remodeling of N-glycans on SLC3A2. Furthermore, proteomics and functional assays revealed an N-glycan-dependent clustering of the SLC3A2∗SLC7A5 heterodimer with amino-acid/Na+ symporters (SLC1A4, SLC1A5) that balances branched-chain amino acids and Gln levels, at the expense of ATP to maintain the Na+/K+ gradient. In replete conditions, SLC3A2 interactions require Golgi-modified N-glycans at N365D and N381D, whereas reducing N-glycosylation in the endoplasmic reticulum by fluvastatin treatment promoted the recruitment of CD44 and transporters needed to mitigate stress. Thus, SLC3A2 N-glycosylation and Golgi remodeling of the N-glycans have distinct roles in amino acids import for growth, maintenance, and metabolic stresses.


Assuntos
Cadeia Pesada da Proteína-1 Reguladora de Fusão , Transportador 1 de Aminoácidos Neutros Grandes , Estresse Fisiológico , Humanos , Trifosfato de Adenosina/metabolismo , Aminoácidos/metabolismo , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Galectina 3/metabolismo , Glicosilação , Células HeLa , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Polissacarídeos/metabolismo
5.
Proc Natl Acad Sci U S A ; 115(33): E7748-E7757, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30065114

RESUMO

Approximately one-third of the mammalian proteome is transported from the endoplasmic reticulum-to-Golgi via COPII-coated vesicles. SEC23, a core component of coat protein-complex II (COPII), is encoded by two paralogous genes in vertebrates (Sec23a and Sec23b). In humans, SEC23B deficiency results in congenital dyserythropoietic anemia type-II (CDAII), while SEC23A deficiency results in a skeletal phenotype (with normal red blood cells). These distinct clinical disorders, together with previous biochemical studies, suggest unique functions for SEC23A and SEC23B. Here we show indistinguishable intracellular protein interactomes for human SEC23A and SEC23B, complementation of yeast Sec23 by both human and murine SEC23A/B, and rescue of the lethality of sec23b deficiency in zebrafish by a sec23a-expressing transgene. We next demonstrate that a Sec23a coding sequence inserted into the murine Sec23b locus completely rescues the lethal SEC23B-deficient pancreatic phenotype. We show that SEC23B is the predominantly expressed paralog in human bone marrow, but not in the mouse, with the reciprocal pattern observed in the pancreas. Taken together, these data demonstrate an equivalent function for SEC23A/B, with evolutionary shifts in the transcription program likely accounting for the distinct phenotypes of SEC23A/B deficiency within and across species, a paradigm potentially applicable to other sets of paralogous genes. These findings also suggest that enhanced erythroid expression of the normal SEC23A gene could offer an effective therapeutic approach for CDAII patients.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Eritrócitos/metabolismo , Complexos Multiproteicos/biossíntese , Proteínas de Transporte Vesicular/biossíntese , Anemia Diseritropoética Congênita/genética , Anemia Diseritropoética Congênita/metabolismo , Medula Óssea/metabolismo , Medula Óssea/patologia , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Eritrócitos/patologia , Regulação da Expressão Gênica , Células HEK293 , Humanos , Complexos Multiproteicos/genética , Especificidade da Espécie , Proteínas de Transporte Vesicular/genética
6.
Proc Natl Acad Sci U S A ; 114(21): 5425-5430, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28487484

RESUMO

MicroRNAs (miRNAs) play critical roles in a broad variety of biological processes by inhibiting translation initiation and by destabilizing target mRNAs. The CCR4-NOT complex effects miRNA-mediated silencing, at least in part through interactions with 4E-T (eIF4E transporter) protein, but the precise mechanism is unknown. Here we show that the cap-binding eIF4E-homologous protein 4EHP is an integral component of the miRNA-mediated silencing machinery. We demonstrate that the cap-binding activity of 4EHP contributes to the translational silencing by miRNAs through the CCR4-NOT complex. Our results show that 4EHP competes with eIF4E for binding to 4E-T, and this interaction increases the affinity of 4EHP for the cap. We propose a model wherein the 4E-T/4EHP interaction engenders a closed-loop mRNA conformation that blocks translational initiation of miRNA targets.


Assuntos
MicroRNAs/metabolismo , Proteínas de Ligação ao Cap de RNA/metabolismo , Interferência de RNA , Complexo de Inativação Induzido por RNA/metabolismo , Fator de Iniciação 4E em Eucariotos , Células HEK293 , Células HeLa , Humanos , Proteínas de Transporte Nucleocitoplasmático/metabolismo
7.
J Cell Sci ; 130(5): 827-840, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28096472

RESUMO

In this study, we demonstrate myosin VI enrichment at Cx43 (also known as GJA1)-containing gap junctions (GJs) in heart tissue, primary cardiomyocytes and cell culture models. In primary cardiac tissue and in fibroblasts from the myosin VI-null mouse as well as in tissue culture cells transfected with siRNA against myosin VI, we observe reduced GJ plaque size with a concomitant reduction in intercellular communication, as shown by fluorescence recovery after photobleaching (FRAP) and a new method of selective calcein administration. Analysis of the molecular role of myosin VI in Cx43 trafficking indicates that myosin VI is dispensable for the delivery of Cx43 to the cell surface and connexon movement in the plasma membrane. Furthermore, we cannot corroborate clathrin or Dab2 localization at gap junctions and we do not observe a function for the myosin-VI-Dab2 complex in clathrin-dependent endocytosis of annular gap junctions. Instead, we found that myosin VI was localized at the edge of Cx43 plaques by using total internal reflection fluorescence (TIRF) microscopy and use FRAP to identify a plaque accretion defect as the primary manifestation of myosin VI loss in Cx43 homeostasis. A fuller understanding of this derangement may explain the cardiomyopathy or gliosis associated with the loss of myosin VI.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/metabolismo , Miocárdio/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Motivos de Aminoácidos , Animais , Comunicação Celular , Membrana Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Complexo de Golgi/metabolismo , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Cadeias Pesadas de Miosina/química , Domínios Proteicos , Transporte Proteico , Ratos
8.
Prog Mol Subcell Biol ; 57: 151-180, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30097775

RESUMO

In addition to being the terminal degradative compartment of the cell's endocytic and autophagic pathways, the lysosome is a multifunctional signalling hub integrating the cell's response to nutrient status and growth factor/hormone signalling. The cytosolic surface of the limiting membrane of the lysosome is the site of activation of the multiprotein complex mammalian target of rapamycin complex 1 (mTORC1), which phosphorylates numerous cell growth-related substrates, including transcription factor EB (TFEB). Under conditions in which mTORC1 is inhibited including starvation, TFEB becomes dephosphorylated and translocates to the nucleus where it functions as a master regulator of lysosome biogenesis. The signalling role of lysosomes is not limited to this pathway. They act as an intracellular Ca2+ store, which can release Ca2+ into the cytosol for both local effects on membrane fusion and pleiotropic effects within the cell. The relationship and crosstalk between the lysosomal and endoplasmic reticulum (ER) Ca2+ stores play a role in shaping intracellular Ca2+ signalling. Lysosomes also perform other signalling functions, which are discussed. Current views of the lysosomal compartment recognize its dynamic nature. It includes endolysosomes, autolysosome and storage lysosomes that are constantly engaged in fusion/fission events and lysosome regeneration. How signalling is affected by individual lysosomal organelles being at different stages of these processes and/or at different sites within the cell is poorly understood, but is discussed.


Assuntos
Endocitose/genética , Retículo Endoplasmático/genética , Endossomos/genética , Lisossomos/genética , Animais , Autofagia/genética , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Humanos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Transdução de Sinais/genética
9.
Glycobiology ; 27(7): 595-598, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29048482

RESUMO

Pedersen et al. (Pedersen HK, Gudmundsdottir V, Nielsen HB, Hyotylainen T, Nielsen T, Jensen BA, Forslund K, Hildebrand F, Prifti E, Falony G, et al. 2016. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature. 535: 376-381.) report that human serum levels of branched-chain amino acids (BCAA) and N-acetylglucosamine (GlcNAc) increase in proportion to insulin resistance. They focus on the microbiome and the contributing subset of microbe species, thereby demonstrating disease causality in mice. As either oral GlcNAc or BCAA in mice are known to increase insulin resistance and weight gain, we note that recently published molecular data argues for a cooperative interaction.


Assuntos
Resistência à Insulina , Acetilglucosamina , Aminoácidos de Cadeia Ramificada , Animais , Humanos , Insulina , Metaboloma , Camundongos
10.
Traffic ; 15(6): 684-99, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24612377

RESUMO

Altered phosphorylation and trafficking of connexin 43 (Cx43) during acute ischemia contributes to arrhythmogenic gap junction remodeling, yet the critical sequence and accessory proteins necessary for Cx43 internalization remain unresolved. 14-3-3 proteins can regulate protein trafficking, and a 14-3-3 mode-1 binding motif is activated upon phosphorylation of Ser373 of the Cx43 C-terminus. We hypothesized that Cx43(Ser373) phosphorylation is important to pathological gap junction remodeling. Immunofluorescence in human heart reveals the enrichment of 14-3-3 proteins at intercalated discs, suggesting interaction with gap junctions. Knockdown of 14-3-3τ in cell lines increases gap junction plaque size at cell-cell borders. Cx43(S373A) mutation prevents Cx43/14-3-3 complexing and stabilizes Cx43 at the cell surface, indicating avoidance of degradation. Using Langendorff-perfused mouse hearts, we detect phosphorylation of newly internalized Cx43 at Ser373 and Ser368 within 30 min of no-flow ischemia. Phosphorylation of Cx43 at Ser368 by protein kinase C and Ser255 by mitogen-activated protein kinase has previously been implicated in Cx43 internalization. The Cx43(S373A) mutant is resistant to phosphorylation at both these residues and does not undergo ubiquitination, revealing Ser373 phosphorylation as an upstream gatekeeper of a posttranslational modification cascade necessary for Cx43 internalization. Cx43(Ser373) phosphorylation is a potent target for therapeutic interventions to preserve gap junction coupling in the stressed myocardium.


Assuntos
Proteínas 14-3-3/metabolismo , Conexina 43/metabolismo , Endocitose , Junções Comunicantes/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Proteínas 14-3-3/química , Proteínas 14-3-3/genética , Motivos de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Conexina 43/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Fosforilação , Ligação Proteica , Proteína Quinase C/metabolismo , Estabilidade Proteica , Transporte Proteico
11.
Circ Res ; 106(6): 1153-63, 2010 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-20167932

RESUMO

RATIONALE: Gap junctions mediate cell-to-cell electric coupling of cardiomyocytes. The primary gap junction protein in the working myocardium, connexin43 (Cx43), exhibits increased localization at the lateral membranes of cardiomyocytes in a variety of heart diseases, although the precise location and function of this population is unknown. OBJECTIVE: To define the subcellular location of lateralized gap junctions at the light and electron microscopic level, and further characterize the biochemical regulation of gap junction turnover. METHODS AND RESULTS: By electron microscopy, we characterized gap junctions formed between cardiomyocyte lateral membranes in failing canine ventricular myocardium. These gap junctions were varied in structure and appeared to be extensively internalizing. Internalized gap junctions were incorporated into multilamellar membrane structures, with features characteristic of autophagosomes. Intracellular Cx43 extensively colocalized with the autophagosome marker GFP-LC3 when both proteins were exogenously expressed in HeLa cells, and endogenous Cx43 colocalized with GFP-LC3 in neonatal rat ventricular myocytes. Furthermore, a distinct phosphorylated form of Cx43, as well as the autophagosome-targeted form of LC3 (microtubule-associated protein light chain 3) targeted to lipid rafts in cardiac tissue, and both were increased in heart failure. CONCLUSIONS: Our data demonstrate a previously unrecognized pathway of gap junction internalization and degradation in the heart and identify a cellular pathway with potential therapeutic implications.


Assuntos
Conexina 43/metabolismo , Junções Comunicantes/metabolismo , Junções Comunicantes/ultraestrutura , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Animais , Autofagia , Conexina 43/genética , Modelos Animais de Doenças , Cães , Células HeLa , Ventrículos do Coração/metabolismo , Ventrículos do Coração/ultraestrutura , Humanos , Microdomínios da Membrana/metabolismo , Microdomínios da Membrana/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
12.
Life Sci Alliance ; 5(11)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35794005

RESUMO

V-ATPases are rotary proton pumps that serve as signaling hubs with numerous protein binding partners. CryoEM with exhaustive focused classification allowed detection of endogenous proteins associated with porcine kidney V-ATPase. An extra C subunit was found in ∼3% of complexes, whereas ∼1.6% of complexes bound mEAK-7, a protein with proposed roles in dauer formation in nematodes and mTOR signaling in mammals. High-resolution cryoEM of porcine kidney V-ATPase with recombinant mEAK-7 showed that mEAK-7's TLDc domain interacts with V-ATPase's stator, whereas its C-terminal α helix binds V-ATPase's rotor. This crosslink would be expected to inhibit rotary catalysis. However, unlike the yeast TLDc protein Oxr1p, exogenous mEAK-7 does not inhibit V-ATPase and mEAK-7 overexpression in cells does not alter lysosomal or phagosomal pH. Instead, cryoEM suggests that the mEAK-7:V-ATPase interaction is disrupted by ATP-induced rotation of the rotor. Comparison of Oxr1p and mEAK-7 binding explains this difference. These results show that V-ATPase binding by TLDc domain proteins can lead to effects ranging from strong inhibition to formation of labile interactions that are sensitive to the enzyme's activity.


Assuntos
ATPases Vacuolares Próton-Translocadoras , Animais , Microscopia Crioeletrônica , Mamíferos/metabolismo , Ligação Proteica , Subunidades Proteicas/química , Suínos , ATPases Vacuolares Próton-Translocadoras/química , ATPases Vacuolares Próton-Translocadoras/metabolismo
13.
Nat Commun ; 13(1): 3812, 2022 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-35780247

RESUMO

Autophagy selectively targets cargo for degradation, yet mechanistic understanding remains incomplete. The ATG8-family plays key roles in autophagic cargo recruitment. Here by mapping the proximal interactome of ATG8-paralogs, LC3B and LC3C, we uncover a LC3C-Endocytic-Associated-Pathway (LEAP) that selectively recruits plasma-membrane (PM) cargo to autophagosomes. We show that LC3C localizes to peripheral endosomes and engages proteins that traffic between PM, endosomes and autophagosomes, including the SNARE-VAMP3 and ATG9, a transmembrane protein essential for autophagy. We establish that endocytic LC3C binds cargo internalized from the PM, including the Met receptor tyrosine kinase and transferrin receptor, and is necessary for their recruitment into ATG9 vesicles targeted to sites of autophagosome initiation. Structure-function analysis identified that LC3C-endocytic localization and engagement with PM-cargo requires the extended carboxy-tail unique to LC3C, the TBK1 kinase, and TBK1-phosphosites on LC3C. These findings identify LEAP as an unexpected LC3C-dependent pathway, providing new understanding of selective coupling of PM signalling with autophagic degradation.


Assuntos
Endossomos , Proteínas Associadas aos Microtúbulos , Autofagia/fisiologia , Membrana Celular/metabolismo , Endossomos/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas SNARE/metabolismo
14.
Circ Res ; 104(3): 355-64, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19122180

RESUMO

Previous studies have postulated an important role for the inwardly rectifying potassium current (I(K1)) in controlling the dynamics of electrophysiological spiral waves responsible for ventricular tachycardia and fibrillation. In this study, we developed a novel tissue model of cultured neonatal rat ventricular myocytes (NRVMs) with uniform or heterogeneous Kir2.1expression achieved by lentiviral transfer to elucidate the role of I(K1) in cardiac arrhythmogenesis. Kir2.1-overexpressed NRVMs showed increased I(K1) density, hyperpolarized resting membrane potential, and increased action potential upstroke velocity compared with green fluorescent protein-transduced NRVMs. Opposite results were observed in Kir2.1-suppressed NRVMs. Optical mapping of uniformly Kir2.1 gene-modified monolayers showed altered conduction velocity and action potential duration compared with nontransduced and empty vector-transduced monolayers, but functional reentrant waves could not be induced. In monolayers with an island of altered Kir2.1 expression, conduction velocity and action potential duration of the locally transduced and nontransduced regions were similar to those of the uniformly transduced and nontransduced monolayers, respectively, and functional reentrant waves could be induced. The waves were anchored to islands of Kir2.1 overexpression and remained stable but dropped in frequency and meandered away from islands of Kir2.1 suppression. In monolayers with an inverse pattern of I(K1) heterogeneity, stable high frequency spiral waves were present with I(K1) overexpression, whereas lower frequency, meandering spiral waves were observed with I(K1) suppression. Our study provides direct evidence for the contribution of I(K1) heterogeneity and level to the genesis and stability of spiral waves and highlights the potential importance of I(K1) as an antiarrhythmia target.


Assuntos
Miócitos Cardíacos/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/genética , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/fisiopatologia , Animais , Células Cultivadas , Heterogeneidade Genética , Proteínas de Fluorescência Verde/genética , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos
15.
J Cell Biol ; 220(2)2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33464297

RESUMO

Adaptor protein complex 5 (AP-5) and its partners, SPG11 and SPG15, are recruited onto late endosomes and lysosomes. Here we show that recruitment of AP-5/SPG11/SPG15 is enhanced in starved cells and occurs by coincidence detection, requiring both phosphatidylinositol 3-phosphate (PI3P) and Rag GTPases. PI3P binding is via the SPG15 FYVE domain, which, on its own, localizes to early endosomes. GDP-locked RagC promotes recruitment of AP-5/SPG11/SPG15, while GTP-locked RagA prevents its recruitment. Our results uncover an interplay between AP-5/SPG11/SPG15 and the mTORC1 pathway and help to explain the phenotype of AP-5/SPG11/SPG15 deficiency in patients, including the defect in autophagic lysosome reformation.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Complexos Multiproteicos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas/metabolismo , Proteínas de Transporte/química , Endossomos/metabolismo , Células HEK293 , Células HeLa , Humanos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Modelos Biológicos , Nucleotídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Domínios Proteicos
16.
J Mol Cell Cardiol ; 48(4): 713-24, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19961855

RESUMO

Increased cyclic GMP from enhanced synthesis or suppressed catabolism (e.g. PDE5 inhibition by sildenafil, SIL) activates protein kinase G (PKG) and blunts cardiac pathological hypertrophy. Suppressed calcineurin (Cn)-NFAT (nuclear factor of activated T-cells) signaling appears to be involved, though it remains unclear how this is achieved. One potential mechanism involves activation of Cn/NFAT by calcium entering via transient receptor potential canonical (TRPC) channels (notably TRPC6). Here, we tested the hypothesis that PKG blocks Cn/NFAT activation by modifying and thus inhibiting TRPC6 current to break the positive feedback loop involving NFAT and NFAT-dependent TRPC6 upregulation. TRPC6 expression rose with pressure-overload in vivo, and angiotensin (ATII) or endothelin (ET1) stimulation in neonatal and adult cardiomyocytes in vitro. 8Br-cGMP and SIL reduced ET1-stimulated TRPC6 expression and NFAT dephosphorylation (activity). TRPC6 upregulation was absent if its promoter was mutated with non-functional NFAT binding sites, whereas constitutively active NFAT triggered TRPC6 expression that was not inhibited by SIL. PKG phosphorylated TRPC6, and both T70 and S322 were targeted. Both sites were functionally relevant, as 8Br-cGMP strongly suppressed current in wild-type TRPC6 channels, but not in those with phospho-silencing mutations (T70A, S322A or S322Q). NFAT activation and increased protein synthesis stimulated by ATII or ET1 was blocked by 8Br-cGMP or SIL. However, transfection with T70A or S322Q TRPC6 mutants blocked this inhibitory effect, whereas phospho-mimetic mutants (T70E, S322E, and both combined) suppressed NFAT activation. Thus PDE5-inhibition blocks TRPC6 channel activation and associated Cn/NFAT activation signaling by PKG-dependent channel phosphorylation.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Regulação da Expressão Gênica , Fatores de Transcrição NFATC/metabolismo , Inibidores da Fosfodiesterase 5 , Canais de Cátion TRPC/metabolismo , Angiotensinas/biossíntese , Animais , Aorta/patologia , Endotelinas/biossíntese , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Células Musculares/citologia , Mutação , Miócitos Cardíacos/citologia , Fosforilação , Piperazinas/farmacologia , Purinas/farmacologia , Ratos , Citrato de Sildenafila , Sulfonas/farmacologia , Canal de Cátion TRPC6
17.
Circulation ; 119(9): 1220-30, 2009 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-19237662

RESUMO

BACKGROUND: Cardiac resynchronization therapy (CRT) is widely applied in patients with heart failure and dyssynchronous contraction (DHF), but the electrophysiological consequences of CRT in heart failure remain largely unexplored. METHODS AND RESULTS: Adult dogs underwent left bundle-branch ablation and either right atrial pacing (190 to 200 bpm) for 6 weeks (DHF) or 3 weeks of right atrial pacing followed by 3 weeks of resynchronization by biventricular pacing at the same pacing rate (CRT). Isolated left ventricular anterior and lateral myocytes from nonfailing (control), DHF, and CRT dogs were studied with the whole-cell patch clamp. Quantitative polymerase chain reaction and Western blots were performed to measure steady state mRNA and protein levels. DHF significantly reduced the inward rectifier K(+) current (I(K1)), delayed rectifier K(+) current (I(K)), and transient outward K(+) current (I(to)) in both anterior and lateral cells. CRT partially restored the DHF-induced reduction of I(K1) and I(K) but not I(to), consistent with trends in the changes in steady state K(+) channel mRNA and protein levels. DHF reduced the peak inward Ca(2+) current (I(Ca)) density and slowed I(Ca) decay in lateral compared with anterior cells, whereas CRT restored peak I(Ca) amplitude but did not hasten decay in lateral cells. Calcium transient amplitudes were depressed and the decay was slowed in DHF, especially in lateral myocytes. CRT hastened the decay in both regions and increased the calcium transient amplitude in lateral but not anterior cells. No difference was found in Ca(V)1.2 (alpha1C) mRNA or protein expression, but reduced Ca(V)beta2 mRNA was found in DHF cells. DHF reduced phospholamban, ryanodine receptor, and sarcoplasmic reticulum Ca(2+) ATPase and increased Na(+)-Ca(2+) exchanger mRNA and protein. CRT did not restore the DHF-induced molecular remodeling, except for sarcoplasmic reticulum Ca(2+) ATPase. Action potential durations were significantly prolonged in DHF, especially in lateral cells, and CRT abbreviated action potential duration in lateral but not anterior cells. Early afterdepolarizations were more frequent in DHF than in control cells and were reduced with CRT. CONCLUSIONS: CRT partially restores DHF-induced ion channel remodeling and abnormal Ca(2+) homeostasis and attenuates the regional heterogeneity of action potential duration. The electrophysiological changes induced by CRT may suppress ventricular arrhythmias, contribute to the survival benefit of this therapy, and improve the mechanical performance of the heart.


Assuntos
Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/fisiologia , Marca-Passo Artificial , Potenciais de Ação/fisiologia , Animais , Bloqueio de Ramo/diagnóstico , Bloqueio de Ramo/fisiopatologia , Bloqueio de Ramo/terapia , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Circulação Coronária , Cães , Ecocardiografia , Eletrocardiografia , Insuficiência Cardíaca/diagnóstico por imagem , Homeostase/fisiologia , Proteínas Interatuantes com Canais de Kv/genética , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , RNA Mensageiro/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo
18.
Science ; 370(6514): 351-356, 2020 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-33060361

RESUMO

The mechanistic target of rapamycin complex 1 (mTORC1) couples nutrient sufficiency to cell growth. mTORC1 is activated by exogenously acquired amino acids sensed through the GATOR-Rag guanosine triphosphatase (GTPase) pathway, or by amino acids derived through lysosomal degradation of protein by a poorly defined mechanism. Here, we revealed that amino acids derived from the degradation of protein (acquired through oncogenic Ras-driven macropinocytosis) activate mTORC1 by a Rag GTPase-independent mechanism. mTORC1 stimulation through this pathway required the HOPS complex and was negatively regulated by activation of the GATOR-Rag GTPase pathway. Therefore, distinct but functionally coordinated pathways control mTORC1 activity on late endocytic organelles in response to distinct sources of amino acids.


Assuntos
Aminoácidos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Lisossomos/enzimologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas R-SNARE/metabolismo , Ativação Enzimática , Células HEK293 , Humanos , Pinocitose , Proteólise
19.
J Cardiovasc Pharmacol ; 54(4): 263-72, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19701097

RESUMO

Gap junctions (GJs) allow direct communication between cells. In the heart, GJs mediate the electrical coupling of cardiomyocytes and as such dictate the speed and direction of cardiac conduction. A prominent feature of acquired structural heart disease is remodeling of GJ protein expression and localization concomitant with increased susceptibility to lethal arrhythmias, leading many to hypothesize that the two are causally linked. Detailed understanding of the cellular mechanisms that regulate GJ localization and function within cardiomyocytes may therefore uncover potential therapeutic strategies for a significant clinical problem. This review will outline our current understanding of GJ cell biology with the intent of highlighting cellular mechanisms responsible for GJ remodeling associated with cardiac disease.


Assuntos
Junções Comunicantes/fisiologia , Cardiopatias/etiologia , Animais , Comunicação Celular/fisiologia , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Junções Comunicantes/ultraestrutura , Sistema de Condução Cardíaco/fisiologia , Cardiopatias/metabolismo , Cardiopatias/fisiopatologia , Humanos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , Fosforilação , Domínios e Motivos de Interação entre Proteínas
20.
J Mol Biol ; 430(14): 2153-2163, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29778605

RESUMO

Multi-subunit tethering complexes control membrane fusion events in eukaryotic cells. Class C core vacuole/endosome tethering (CORVET) and homotypic fusion and vacuole protein sorting (HOPS) are two such complexes, both containing the Sec1/Munc18 protein subunit VPS33A. Metazoans additionally possess VPS33B, which has considerable sequence similarity to VPS33A but does not integrate into CORVET or HOPS complexes and instead stably interacts with VIPAR. It has been recently suggested that VPS33B and VIPAR comprise two subunits of a novel multi-subunit tethering complex (named "CHEVI"), perhaps analogous in configuration to CORVET and HOPS. We utilized the BioID proximity biotinylation assay to compare and contrast the interactomes of VPS33A and VPS33B. Overall, few proteins were identified as associating with both VPS33A and VPS33B, suggesting that these proteins have distinct sub-cellular localizations. Consistent with previous reports, we observed that VPS33A was co-localized with many components of class III phosphatidylinositol 3-kinase (PI3KC3) complexes: PIK3C3, PIK3R4, NRBF2, UVRAG and RUBICON. Although VPS33A clearly co-localized with several subunits of CORVET and HOPS in this assay, no proteins with the canonical CORVET/HOPS domain architecture were found to co-localize with VPS33B. Instead, we identified that VPS33B interacts directly with CCDC22, a member of the CCC complex. CCDC22 does not co-fractionate with VPS33B and VIPAR in gel filtration of human cell lysates, suggesting that CCDC22 interacts transiently with VPS33B/VIPAR rather than forming a stable complex with these proteins in cells. We also observed that the protein complex containing VPS33B and VIPAR is considerably smaller than CORVET/HOPS, suggesting that the CHEVI complex comprises just VPS33B and VIPAR.


Assuntos
Proteômica/métodos , Proteínas de Transporte Vesicular/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Modelos Moleculares , Fosfatidilinositol 3-Quinase/metabolismo , Conformação Proteica , Mapeamento de Interação de Proteínas , Proteínas/metabolismo , Células THP-1 , Proteínas de Transporte Vesicular/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA