Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Mol Cell ; 83(14): 2524-2539.e7, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37390818

RESUMEN

Maintaining a highly acidic lysosomal pH is central to cellular physiology. Here, we use functional proteomics, single-particle cryo-EM, electrophysiology, and in vivo imaging to unravel a key biological function of human lysosome-associated membrane proteins (LAMP-1 and LAMP-2) in regulating lysosomal pH homeostasis. Despite being widely used as a lysosomal marker, the physiological functions of the LAMP proteins have long been overlooked. We show that LAMP-1 and LAMP-2 directly interact with and inhibit the activity of the lysosomal cation channel TMEM175, a key player in lysosomal pH homeostasis implicated in Parkinson's disease. This LAMP inhibition mitigates the proton conduction of TMEM175 and facilitates lysosomal acidification to a lower pH environment crucial for optimal hydrolase activity. Disrupting the LAMP-TMEM175 interaction alkalinizes the lysosomal pH and compromises the lysosomal hydrolytic function. In light of the ever-increasing importance of lysosomes to cellular physiology and diseases, our data have widespread implications for lysosomal biology.


Asunto(s)
Enfermedad de Parkinson , Humanos , Concentración de Iones de Hidrógeno , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Enfermedad de Parkinson/metabolismo , Canales de Potasio/metabolismo
2.
Nat Immunol ; 24(1): 136-147, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36581712

RESUMEN

Hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) by phospholipase C-γ (PLCγ1) represents a critical step in T cell antigen receptor (TCR) signaling and subsequent thymocyte and T cell responses. PIP2 replenishment following its depletion in the plasma membrane (PM) is dependent on delivery of its precursor phosphatidylinositol (PI) from the endoplasmic reticulum (ER) to the PM. We show that a PI transfer protein (PITP), Nir3 (Pitpnm2), promotes PIP2 replenishment following TCR stimulation and is important for T cell development. In Nir3-/- T lineage cells, the PIP2 replenishment following TCR stimulation is slower. Nir3 deficiency attenuates calcium mobilization in double-positive (DP) thymocytes in response to weak TCR stimulation. This impaired TCR signaling leads to attenuated thymocyte development at TCRß selection and positive selection as well as diminished mature T cell fitness in Nir3-/- mice. This study highlights the importance of PIP2 replenishment mediated by PITPs at ER-PM junctions during TCR signaling.


Asunto(s)
Proteínas de Transferencia de Fosfolípidos , Transducción de Señal , Ratones , Animales , Proteínas de Transferencia de Fosfolípidos/metabolismo , Membrana Celular/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Fosfatidilinositoles/metabolismo
3.
Biomolecules ; 12(12)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36551225

RESUMEN

Gramicidin A (gA) is a linear antimicrobial peptide that can form a channel and specifically conduct monovalent cations such as H+ across the lipid membrane. The antimicrobial activity of gA is associated with the formation of hydroxyl free radicals and the imbalance of NADH metabolism, possibly a consequence caused by the conductance of cations. The ion conductivity of gramicidin A can be blocked by Ca2+ ions. However, the effect of Ca2+ ions on the antimicrobial activity of gA is unclear. To unveil the role of Ca2+ ions, we examined the effect of Ca2+ ions on the antimicrobial activity of gramicidin A against Staphylococcus aureus (S. aureus). Results showed that the antimicrobial mechanism of gA and antimicrobial activity by Ca2+ ions are concentration-dependent. At the low gA concentration (≤1 µM), the antimicrobial mechanism of gA is mainly associated with the hydroxyl free radical formation and NADH metabolic imbalance. Under this mode, Ca2+ ions can significantly inhibit the hydroxyl free radical formation and NADH metabolic imbalance. On the other hand, at high gA concentration (≥5 µM), gramicidin A acts more likely as a detergent. Gramicidin A not only causes an increase in hydroxyl free radical levels and NAD+/NADH ratios but also induces the destruction of the lipid membrane composition. At this condition, Ca2+ ions can no longer reduce the gA antimicrobial activity but rather enhance the bacterial killing ability of gramicidin A.


Asunto(s)
Antibacterianos , Calcio , Gramicidina , Staphylococcus aureus , Antibacterianos/química , Antibacterianos/farmacología , Calcio/metabolismo , Cationes Bivalentes , Membrana Celular/metabolismo , Gramicidina/química , Gramicidina/farmacología , Lípidos de la Membrana/metabolismo , NAD/metabolismo , Staphylococcus aureus/efectos de los fármacos
4.
Mol Biol Cell ; 33(3): br2, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35020418

RESUMEN

Homeostatic regulation of plasma membrane (PM) phosphatidylinositol 4,5-bisphosphate (PIP2) in receptor-stimulated cells is mediated by the lipid transfer protein Nir2. Nir2 is dynamically recruited to endoplasmic reticulum-plasma membrane (ER-PM) junctions to facilitate replenishment of PM PIP2 hydrolyzed during receptor-mediated signaling. However, our knowledge regarding the activation and sustainment of Nir2-mediated replenishment of PM PIP2 is limited. Here, we describe the functions of Nir1 as a positive regulator of Nir2 and PIP2 homeostasis. In contrast to the family proteins Nir2 and Nir3, Nir1 constitutively localizes at ER-PM junctions. Nir1 potentiates Nir2 targeting to ER-PM junctions during receptor-mediated signaling and is required for efficient PM PIP2 replenishment. Live-cell imaging and biochemical analysis reveal that Nir1 interacts with Nir2 via a region between the FFAT motif and the DDHD domain. Combined, results from this study identify Nir1 as an ER-PM junction localized protein that promotes Nir2 recruitment for PIP2 homeostasis.


Asunto(s)
Retículo Endoplásmico , Proteínas de la Membrana , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Homeostasis , Proteínas de la Membrana/metabolismo , Fosfatidilinositoles/metabolismo
5.
Elife ; 102021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33973848

RESUMEN

Sigma 1 receptor (S1R) is a 223-amino-acid-long transmembrane endoplasmic reticulum (ER) protein. S1R modulates activity of multiple effector proteins and is a well-established drug target. However, signaling functions of S1R in cells are poorly understood. Here, we test the hypothesis that biological activity of S1R in cells can be explained by its ability to interact with cholesterol and to form cholesterol-enriched microdomains in the ER membrane. By performing experiments in reduced reconstitution systems, we demonstrate direct effects of cholesterol on S1R clustering. We identify a novel cholesterol-binding motif in the transmembrane region of human S1R. Mutations of this motif impair association of recombinant S1R with cholesterol beads, affect S1R clustering in vitro and disrupt S1R subcellular localization. We demonstrate that S1R-induced membrane microdomains have increased local membrane thickness and that increased local cholesterol concentration and/or membrane thickness in these microdomains can modulate signaling of inositol-requiring enzyme 1α in the ER. Further, S1R agonists cause disruption of S1R clusters, suggesting that biological activity of S1R agonists is linked to remodeling of ER membrane microdomains. Our results provide novel insights into S1R-mediated signaling mechanisms in cells.


Asunto(s)
Colesterol/metabolismo , Retículo Endoplásmico/metabolismo , Receptores sigma/genética , Receptores sigma/metabolismo , Transducción de Señal , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Microdominios de Membrana , Unión Proteica , Receptor Sigma-1
6.
Science ; 372(6545): 935-941, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33927055

RESUMEN

During infection, intracellular bacterial pathogens translocate a variety of effectors into host cells that modify host membrane trafficking for their benefit. We found a self-organizing system consisting of a bacterial phosphoinositide kinase and its opposing phosphatase that formed spatiotemporal patterns, including traveling waves, to remodel host cellular membranes. The Legionella effector MavQ, a phosphatidylinositol (PI) 3-kinase, was targeted to the endoplasmic reticulum (ER). MavQ and the Legionella PI 3-phosphatase SidP, even in the absence of other bacterial components, drove rapid PI 3-phosphate turnover on the ER and spontaneously formed traveling waves that spread along ER subdomains inducing vesicle and tubule budding. Thus, bacteria can exploit a self-organizing membrane-targeting mechanism to hijack host cellular structures for survival.


Asunto(s)
Proteínas Bacterianas/metabolismo , Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Legionella pneumophila/fisiología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Proteínas Bacterianas/química , Células COS , Chlorocebus aethiops , Retículo Endoplásmico/ultraestructura , Retroalimentación Fisiológica , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Membranas Intracelulares/ultraestructura , Legionella pneumophila/enzimología , Legionella pneumophila/genética , Legionella pneumophila/crecimiento & desarrollo , Ratones , Mutación , Fosfatidilinositol 3-Quinasa/química , Fosfatos de Fosfatidilinositol/química , Monoéster Fosfórico Hidrolasas/metabolismo , Dominios Proteicos , Células RAW 264.7
7.
Contact (Thousand Oaks) ; 4: 25152564211026505, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-37366370

RESUMEN

Recent studies indicated potential importance of membrane contact sites (MCS) between the endoplasmic reticulum (ER) and other cellular organelles. These MCS have unique protein and lipid composition and serve as hubs for inter-organelle communication and signaling. Despite extensive investigation of MCS protein composition and functional roles, little is known about the process of MCS formation. In this perspective, we propose a hypothesis that MCS are formed not as a result of random interactions between membranes of ER and other organelles but on the basis of pre-existing cholesterol-enriched ER microdomains.

8.
Elife ; 92020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32808593

RESUMEN

Pathogens find diverse niches for survival including inside a host cell where replication occurs in a relatively protective environment. Vibrio parahaemolyticus is a facultative intracellular pathogen that uses its type 3 secretion system 2 (T3SS2) to invade and replicate inside host cells. Analysis of the T3SS2 pathogenicity island encoding the T3SS2 appeared to lack a mechanism for egress of this bacterium from the invaded host cell. Using a combination of molecular tools, we found that VPA0226, a constitutively secreted lipase, is required for escape of V. parahaemolyticus from the host cells. This lipase must be delivered into the host cytoplasm where it preferentially uses fatty acids associated with innate immune response to esterify cholesterol, weakening the plasma membrane and allowing egress of the bacteria. This study reveals the resourcefulness of microbes and the interplay between virulence systems and host cell resources to evolve an ingenious scheme for survival and escape.


Asunto(s)
Proteínas Bacterianas/metabolismo , Colesterol/metabolismo , Ácidos Grasos/metabolismo , Lipasa/metabolismo , Vibrio parahaemolyticus/metabolismo , Esterificación , Islas Genómicas , Sistemas de Secreción Tipo III , Vibrio parahaemolyticus/enzimología
9.
Proc Natl Acad Sci U S A ; 117(2): 993-999, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31879354

RESUMEN

An intimate link exists between circadian clocks and metabolism with nearly every metabolic pathway in the mammalian liver under circadian control. Circadian regulation of metabolism is largely driven by rhythmic transcriptional activation of clock-controlled genes. Among these output genes, Nocturnin (Noct) has one of the highest amplitude rhythms at the mRNA level. The Noct gene encodes a protein (NOC) that is highly conserved with the endonuclease/exonuclease/phosphatase (EEP) domain-containing CCR4 family of deadenylases, but highly purified NOC possesses little or no ribonuclease activity. Here, we show that NOC utilizes the dinucleotide NADP(H) as a substrate, removing the 2' phosphate to generate NAD(H), and is a direct regulator of oxidative stress response through its NADPH 2' phosphatase activity. Furthermore, we describe two isoforms of NOC in the mouse liver. The cytoplasmic form of NOC is constitutively expressed and associates externally with membranes of other organelles, including the endoplasmic reticulum, via N-terminal glycine myristoylation. In contrast, the mitochondrial form of NOC possesses high-amplitude circadian rhythmicity with peak expression level during the early dark phase. These findings suggest that NOC regulates local intracellular concentrations of NADP(H) in a manner that changes over the course of the day.


Asunto(s)
Ritmo Circadiano/fisiología , Hígado/metabolismo , Proteínas Nucleares/metabolismo , Nucleotidasas/metabolismo , Estrés Oxidativo/fisiología , Factores de Transcripción/metabolismo , Animales , Relojes Circadianos/genética , Relojes Circadianos/fisiología , Ritmo Circadiano/genética , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas Nucleares/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Transcriptoma
11.
Elife ; 82019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-31535977

RESUMEN

RET is a receptor tyrosine kinase (RTK) that plays essential roles in development and has been implicated in several human diseases. Different from most of RTKs, RET requires not only its cognate ligands but also co-receptors for activation, the mechanisms of which remain unclear due to lack of high-resolution structures of the ligand/co-receptor/receptor complexes. Here, we report cryo-EM structures of the extracellular region ternary complexes of GDF15/GFRAL/RET, GDNF/GFRα1/RET, NRTN/GFRα2/RET and ARTN/GFRα3/RET. These structures reveal that all the four ligand/co-receptor pairs, while using different atomic interactions, induce a specific dimerization mode of RET that is poised to bring the two kinase domains into close proximity for cross-phosphorylation. The NRTN/GFRα2/RET dimeric complex further pack into a tetrameric assembly, which is shown by our cell-based assays to regulate the endocytosis of RET. Our analyses therefore reveal both the common mechanism and diversification in the activation of RET by different ligands.


Asunto(s)
Activación Enzimática , Proteínas Proto-Oncogénicas c-ret/química , Proteínas Proto-Oncogénicas c-ret/metabolismo , Microscopía por Crioelectrón , Factor Neurotrófico Derivado de la Línea Celular Glial/química , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor 15 de Diferenciación de Crecimiento/química , Factor 15 de Diferenciación de Crecimiento/metabolismo , Humanos , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Neurturina/química , Neurturina/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Procesamiento Proteico-Postraduccional
12.
Biochemistry ; 58(25): 2809-2813, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31184863

RESUMEN

An early step in signaling from activated receptor tyrosine kinases (RTKs) is the recruitment of cytosolic adaptor proteins to autophosphorylated tyrosines in the receptor cytoplasmic domains. Fibroblast growth factor receptor substrate 2α (FRS2α) associates via its phosphotyrosine-binding domain (PTB) to FGF receptors (FGFRs). Upon FGFR activation, FRS2α undergoes phosphorylation on multiple tyrosines, triggering recruitment of the adaptor Grb2 and the tyrosine phosphatase Shp2, resulting in stimulation of PI3K/AKT and MAPK signaling pathways. FRS2α also undergoes N-myristoylation, which was shown to be important for its localization to membranes and its ability to stimulate downstream signaling events (Kouhara et al., 1997). Here we show that FRS2α is also palmitoylated in cells and that cysteines 4 and 5 account for the entire modification. We further show that mutation of those two cysteines interferes with FRS2α localization to the plasma membrane (PM), and we quantify this observation using fluorescence fluctuation spectroscopy approaches. Importantly, prevention of myristoylation by introduction of a G2A mutation also abrogates palmitoylation, raising the possibility that signaling defects previously ascribed to the G2A mutant may actually be due to a failure of that mutant to undergo palmitoylation. Our results demonstrate that FRS2α undergoes coupled myristoylation and palmitoylation. Unlike stable cotranslational modifications, such as myristoylation and prenylation, palmitoylation is reversible due to the relative lability of the thioester linkage. Therefore, palmitoylation may provide a mechanism, in addition to phosphorylation, for dynamic regulation of FRS2 and its downstream signaling pathways.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Lipoilación/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular Tumoral , Cisteína/química , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mutación , Ácido Mirístico/metabolismo , Ácido Palmítico/metabolismo , Espectrometría de Fluorescencia
13.
J Exp Med ; 216(4): 867-883, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30886058

RESUMEN

STING gain-of-function mutations cause lung disease and T cell cytopenia through unknown mechanisms. Here, we found that these mutants induce chronic activation of ER stress and unfolded protein response (UPR), leading to T cell death by apoptosis in the StingN153S/+ mouse and in human T cells. Mechanistically, STING-N154S disrupts calcium homeostasis in T cells, thus intrinsically primes T cells to become hyperresponsive to T cell receptor signaling-induced ER stress and the UPR, leading to cell death. This intrinsic priming effect is mediated through a novel region of STING that we name "the UPR motif," which is distinct from known domains required for type I IFN signaling. Pharmacological inhibition of ER stress prevented StingN153S/+ T cell death in vivo. By crossing StingN153S/+ to the OT-1 mouse, we fully restored CD8+ T cells and drastically ameliorated STING-associated lung disease. Together, our data uncover a critical IFN-independent function of STING that regulates calcium homeostasis, ER stress, and T cell survival.


Asunto(s)
Apoptosis/genética , Linfocitos T CD8-positivos/metabolismo , Calcio/metabolismo , Estrés del Retículo Endoplásmico/genética , Homeostasis/genética , Proteínas de la Membrana/metabolismo , Animales , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Mutación con Ganancia de Función , Células HEK293 , Humanos , Enfermedades Pulmonares/metabolismo , Activación de Linfocitos/genética , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Transfección , Respuesta de Proteína Desplegada/genética
14.
Methods Mol Biol ; 1949: 1-11, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30790244

RESUMEN

Phosphatidylinositol (PI) is an inositol-containing phospholipid synthesized in the endoplasmic reticulum (ER). PI is a precursor lipid for PI 4,5-bisphosphate (PI(4,5)P2) in the plasma membrane (PM) important for Ca2+ signaling in response to extracellular stimuli. Thus, ER-to-PM PI transfer becomes essential for cells to maintain PI(4,5)P2 homeostasis during receptor stimulation. In this chapter, we discuss two live-cell imaging protocols to analyze ER-to-PM PI transfer at ER-PM junctions, where the two membrane compartments make close appositions accommodating PI transfer. First, we describe how to monitor PI(4,5)P2 replenishment following receptor stimulation, as a readout of PI transfer, using a PI(4,5)P2 biosensor and total internal reflection fluorescence microscopy. The second protocol directly visualizes PI transfer proteins that accumulate at ER-PM junctions and mediate PI(4,5)P2 replenishment with PI in the ER in stimulated cells. These methods provide spatial and temporal analysis of ER-to-PM PI transfer during receptor stimulation and can be adapted to other research questions related to this topic.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Fosfatidilinositoles/metabolismo , Transporte Biológico , Técnicas Biosensibles , Células HeLa , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía , Fosfatidilinositol 4,5-Difosfato/metabolismo
15.
Curr Opin Cell Biol ; 57: 99-105, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30739879

RESUMEN

ER-PM junctions are subcellular sites where the endoplasmic reticulum (ER) and the plasma membrane (PM) are kept in close appositions, providing a platform for inter-organelle contact. These membrane contact sites are important for many physiological functions in mammalian cells, including excitation-contraction coupling, store-operated Ca2+ entry, and non-vesicular transfer of lipids between the ER and the PM. Here we review recent insights into the 3D structure and spatial organization of ER-PM junctions in mammalian cells as well as molecular mechanisms underlying the formation and functions of mammalian ER-PM junctions.


Asunto(s)
Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Animales , Calcio/metabolismo , Humanos , Metabolismo de los Lípidos , Mamíferos , Proteínas de la Membrana/metabolismo
16.
Sci Rep ; 8(1): 10477, 2018 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-29992992

RESUMEN

FARP1 is a multi-domain protein that is involved in regulating neuronal development through interacting with cell surface proteins such as class A Plexins and SynCAM 1. The N-terminal FERM domain in FARP1 is known to both promote membrane localization and mediate these protein interactions, for which the underlying molecular mechanisms remain unclear. Here we determined the crystal structures of the FERM domain of FARP1 from zebrafish, and those of FARP2 (a close homolog of FARP1) from mouse and zebrafish. These FERM domains adopt the three-leaved clover fold that is typical of all FERM domains. Our structures reveal a positively charged surface patch that is highly conserved in the FERM domain of FARP1 and FARP2. In vitro lipid-binding experiments showed that the FARP1 FERM domain binds specifically to several types of phospholipid, which is dependent on the positively charged surface patch. We further determined through cell-based analyses that this surface patch on the FERM domain underlies the localization of FARP1 to the plasma membrane, and that FERM domain interactions recruit it to postsynaptic sites in neurons.


Asunto(s)
Membrana Celular/metabolismo , Dominios FERM , Factores de Intercambio de Guanina Nucleótido Rho/química , Pez Cebra/metabolismo , Animales , Sitios de Unión , Cristalografía por Rayos X , Neuronas/metabolismo , Fosfolípidos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína
17.
J Cell Biol ; 217(6): 2047-2058, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29563214

RESUMEN

The endoplasmic reticulum (ER) Ca2+ sensor STIM1 forms oligomers and translocates to ER-plasma membrane (PM) junctions to activate store-operated Ca2+ entry (SOCE) after ER Ca2+ depletion. STIM1 also interacts with EB1 and dynamically tracks microtubule (MT) plus ends. Nevertheless, the role of STIM1-EB1 interaction in regulating SOCE remains unresolved. Using live-cell imaging combined with a synthetic construct approach, we found that EB1 binding constitutes a trapping mechanism restricting STIM1 targeting to ER-PM junctions. We further showed that STIM1 oligomers retain EB1 binding ability in ER Ca2+-depleted cells. By trapping STIM1 molecules at dynamic contacts between the ER and MT plus ends, EB1 binding delayed STIM1 translocation to ER-PM junctions during ER Ca2+ depletion and prevented excess SOCE and ER Ca2+ overload. Our study suggests that STIM1-EB1 interaction shapes the kinetics and amplitude of local SOCE in cellular regions with growing MTs and contributes to spatiotemporal regulation of Ca2+ signaling crucial for cellular functions and homeostasis.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Secuencia de Aminoácidos , Canales de Calcio , Células HeLa , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Proteína ORAI1/metabolismo , Unión Proteica , Transporte de Proteínas , Molécula de Interacción Estromal 1/química
18.
Mol Biol Cell ; 28(23): 3171-3180, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-28954864

RESUMEN

Endoplasmic reticulum-plasma membrane (ER-PM) junctions mediate crucial activities ranging from Ca2+ signaling to lipid metabolism. Spatial organization of ER-PM junctions may modulate the extent and location of these cellular activities. However, the morphology and distribution of ER-PM junctions are not well characterized. Using photoactivated localization microscopy, we reveal that the contact area of single ER-PM junctions is mainly oblong with the dimensions of ∼120 nm × âˆ¼80 nm in HeLa cells. Using total internal reflection fluorescence microscopy and structure illumination microscopy, we show that cortical actin contributes to spatial distribution and stability of ER-PM junctions. Further functional assays suggest that intact F-actin architecture is required for phosphatidylinositol 4,5-bisphosphate homeostasis mediated by Nir2 at ER-PM junctions. Together, our study provides quantitative information on spatial organization of ER-PM junctions that is in part regulated by F-actin. We envision that functions of ER-PM junctions can be differentially regulated through dynamic actin remodeling during cellular processes.


Asunto(s)
Actinas/metabolismo , Retículo Endoplásmico/metabolismo , Análisis Espacial , Citoesqueleto de Actina/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Retículo Endoplásmico/fisiología , Células HeLa , Homeostasis , Humanos , Proteínas de la Membrana/metabolismo , Fosfatidilinositoles/metabolismo , Transducción de Señal
19.
J Cell Biol ; 216(7): 2011-2025, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28600435

RESUMEN

RAS association domain family 4 (RASSF4) is involved in tumorigenesis and regulation of the Hippo pathway. In this study, we identify new functional roles of RASSF4. First, we discovered that RASSF4 regulates store-operated Ca2+ entry (SOCE), a fundamental Ca2+ signaling mechanism, by affecting the translocation of the endoplasmic reticulum (ER) Ca2+ sensor stromal interaction molecule 1 (STIM1) to ER-plasma membrane (PM) junctions. It was further revealed that RASSF4 regulates the formation of ER-PM junctions and the ER-PM tethering function of extended synaptotagmins E-Syt2 and E-Syt3. Moreover, steady-state PM phosphatidylinositol 4,5-bisphosphate (PI[4,5]P2) levels, important for localization of STIM1 and E-Syts at ER-PM junctions, were reduced in RASSF4-knockdown cells. Furthermore, we demonstrated that RASSF4 interacts with and regulates the activity of adenosine diphosphate ribosylation factor 6 (ARF6), a small G protein and upstream regulator of type I phosphatidylinositol phosphate kinases (PIP5Ks) and PM PI(4,5)P2 levels. Overall, our study suggests that RASSF4 controls SOCE and ER-PM junctions through ARF6-dependent regulation of PM PI(4,5)P2 levels, pivotal for a variety of physiological processes.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Femenino , Células HeLa , Humanos , Microscopía Fluorescente , Microscopía por Video , Proteínas de Neoplasias/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transporte de Proteínas , Interferencia de ARN , Molécula de Interacción Estromal 1/genética , Sinaptotagmina II/genética , Sinaptotagmina II/metabolismo , Sinaptotagminas/genética , Sinaptotagminas/metabolismo , Factores de Tiempo , Imagen de Lapso de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
20.
Biochim Biophys Acta Mol Cell Res ; 1864(9): 1494-1506, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28554772

RESUMEN

Endoplasmic reticulum (ER)-plasma membrane (PM) junctions are membrane microdomains important for communication between the ER and the PM. ER-PM junctions were first reported in muscle cells in 1957, but mostly ignored in non-excitable cells due to their scarcity and lack of functional significance. In 2005, the discovery of stromal interaction molecule 1 (STIM1) mediating a universal Ca2+ feedback mechanism at ER-PM junctions in mammalian cells led to a resurgence of research interests toward ER-PM junctions. In the past decade, several major advancements have been made in this emerging topic in cell biology, including the generation of tools for labeling ER-PM junctions and the unraveling of mechanisms underlying regulation and functions of ER-PM junctions. This review summarizes early studies, recently developed tools, and current advances in the characterization and understanding of ER-PM junctions. This article is part of a Special Issue entitled: Membrane Contact Sites edited by Christian Ungermann and Benoit Kornmann.


Asunto(s)
Retículo Endoplásmico/metabolismo , Membranas Intracelulares/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Retículo Endoplásmico/ultraestructura , Humanos , Membranas Intracelulares/ultraestructura , Fusión de Membrana , Fosfolípidos/metabolismo , Transducción de Señal , Esteroles/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA