Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
EMBO Rep ; 25(1): 144-167, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177906

RESUMEN

The tight junction (TJ) in epithelial cells is formed by integral membrane proteins and cytoplasmic scaffolding proteins. The former contains the claudin family proteins with four transmembrane segments, while the latter includes Par3, a PDZ domain-containing adaptor that organizes TJ formation. Here we show the single membrane-spanning protein TMEM25 localizes to TJs in epithelial cells and binds to Par3 via a PDZ-mediated interaction with its C-terminal cytoplasmic tail. TJ development during epithelial cell polarization is accelerated by depletion of TMEM25, and delayed by overexpression of TMEM25 but not by that of a C-terminally deleted protein, indicating a regulatory role of TMEM25. TMEM25 associates via its N-terminal extracellular domain with claudin-1 and claudin-2 to suppress their cis- and trans-oligomerizations, both of which participate in TJ strand formation. Furthermore, Par3 attenuates TMEM25-claudin association via binding to TMEM25, implying its ability to affect claudin oligomerization. Thus, the TJ protein TMEM25 appears to negatively regulate claudin assembly in TJ formation, which regulation is modulated by its interaction with Par3.


Asunto(s)
Claudinas , Uniones Estrechas , Uniones Estrechas/metabolismo , Claudinas/genética , Claudinas/metabolismo , Proteínas Portadoras/metabolismo , Células Epiteliales , Claudina-1/genética , Claudina-1/metabolismo
2.
Genes Cells ; 29(1): 63-72, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985134

RESUMEN

The hydrogen peroxide (H2 O2 )-producing NADPH oxidase Nox4, forming a heterodimer with p22phox , is expressed in a variety of cells including those in the heart to mediate adaptive responses to cellular stresses such as hypoxia. Since Nox4 is constitutively active, H2 O2 production is controlled by its protein abundance. Hypoxia-induced Nox4 expression is observed in various types of cells and generally thought to be regulated at the transcriptional level. Here we show that hypoxia upregulates the Nox4 protein level and Nox4-catalyzed H2 O2 production without increasing the Nox4 mRNA in rat H9c2 cardiomyocytes. In these cells, the Nox4 protein is stabilized under hypoxic conditions in a manner dependent on the presence of p22phox . Cell treatment with the proteasome inhibitor MG132 results in a marked decrease of the Nox4 protein under both normoxic and hypoxic conditions, indicating that the proteasome pathway does not play a major role in Nox4 degradation. The decrease is partially restored by the autophagy inhibitor 3-methyladenine. Furthermore, the Nox4 protein level is upregulated by the lysosome inhibitors bafilomycin A1 and chloroquine. Thus, in cardiomyocytes, Nox4 appears to be degraded via an autophagy-related pathway, and its suppression by hypoxia likely stabilizes Nox4, leading to upregulation of Nox4-catalyzed H2 O2 production.


Asunto(s)
Miocitos Cardíacos , Oxidorreductasas , Ratas , Animales , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Miocitos Cardíacos/metabolismo , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Hipoxia , Autofagia , Especies Reactivas de Oxígeno/metabolismo
3.
J Biol Chem ; 298(10): 102475, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36089063

RESUMEN

The adhesion family of G protein-coupled receptors (GPCRs) is defined by an N-terminal large extracellular region that contains various adhesion-related domains and a highly-conserved GPCR-autoproteolysis-inducing (GAIN) domain, the latter of which is located immediately before a canonical seven-transmembrane domain. These receptors are expressed widely and involved in various functions including development, angiogenesis, synapse formation, and tumorigenesis. GPR125 (ADGRA3), an orphan adhesion GPCR, has been shown to modulate planar cell polarity in gastrulating zebrafish, but its biochemical properties and role in mammalian cells have remained largely unknown. Here, we show that human GPR125 likely undergoes cis-autoproteolysis when expressed in canine kidney epithelial MDCK cells and human embryonic kidney HEK293 cells. The cleavage appears to occur at an atypical GPCR proteolysis site within the GAIN domain during an early stage of receptor biosynthesis. The products, i.e., the N-terminal and C-terminal fragments, seem to remain associated after self-proteolysis, as observed in other adhesion GPCRs. Furthermore, in polarized MDCK cells, GPR125 is exclusively recruited to the basolateral domain of the plasma membrane. The recruitment likely requires the C-terminal PDZ-domain-binding motif of GPR125 and its interaction with the cell polarity protein Dlg1. Knockdown of GPR125 as well as that of Dlg1 results in formation of aberrant cysts with multiple lumens in Matrigel 3D culture of MDCK cells. Consistent with the multilumen phenotype, mitotic spindles are incorrectly oriented during cystogenesis in GPR125-KO MDCK cells. Thus, the basolateral protein GPR125, an autocleavable adhesion GPCR, appears to play a crucial role in apicobasal polarization in epithelial cells.


Asunto(s)
Receptores Acoplados a Proteínas G , Pez Cebra , Animales , Perros , Humanos , Adhesión Celular , Membrana Celular/metabolismo , Polaridad Celular , Homólogo 1 de la Proteína Discs Large/metabolismo , Células HEK293 , Mamíferos/metabolismo , Unión Proteica , Receptores Acoplados a Proteínas G/metabolismo , Pez Cebra/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Secuencias de Aminoácidos
4.
J Biol Chem ; 297(6): 101354, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34717957

RESUMEN

Hepatocytes differ from columnar epithelial cells by their multipolar organization, which follows the initial formation of central lumen-sharing clusters of polarized cells as observed during liver development and regeneration. The molecular mechanism for hepatocyte polarity establishment, however, has been comparatively less studied than those for other epithelial cell types. Here, we show that the tight junction protein Par3 organizes hepatocyte polarization via cooperating with the small GTPase Cdc42 to target atypical protein kinase C (aPKC) to a cortical site near the center of cell-cell contacts. In 3D Matrigel culture of human hepatocytic HepG2 cells, which mimics a process of liver development and regeneration, depletion of Par3, Cdc42, or aPKC results in an impaired establishment of apicobasolateral polarity and a loss of subsequent apical lumen formation. The aPKC activity is also required for bile canalicular (apical) elongation in mouse primary hepatocytes. The lateral membrane-associated proteins Lgl1 and Lgl2, major substrates of aPKC, seem to be dispensable for hepatocyte polarity establishment because Lgl-depleted HepG2 cells are able to form a single apical lumen in 3D culture. On the other hand, Lgl depletion leads to lateral invasion of aPKC, and overexpression of Lgl1 or Lgl2 prevents apical lumen formation, indicating that they maintain proper lateral integrity. Thus, hepatocyte polarity establishment and apical lumen formation are organized by Par3, Cdc42, and aPKC; Par3 cooperates with Cdc42 to recruit aPKC, which plays a crucial role in apical membrane development and regulation of the lateral maintainer Lgl.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/análisis , Proteínas de Ciclo Celular/análisis , Proteínas del Citoesqueleto/análisis , Hepatocitos/citología , Isoenzimas/análisis , Proteína Quinasa C/análisis , Proteína de Unión al GTP cdc42/análisis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Polaridad Celular , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos ICR , Proteína Quinasa C/metabolismo , Proteína de Unión al GTP cdc42/metabolismo
5.
J Biol Chem ; 294(51): 19655-19666, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31732560

RESUMEN

Proper mitotic spindle orientation requires that astral microtubules are connected to the cell cortex by the microtubule-binding protein NuMA, which is recruited from the cytoplasm. Cortical recruitment of NuMA is at least partially mediated via direct binding to the adaptor protein LGN. LGN normally adopts a closed conformation via an intramolecular interaction between its N-terminal NuMA-binding domain and its C-terminal region that contains four GoLoco (GL) motifs, each capable of binding to the membrane-anchored Gαi subunit of heterotrimeric G protein. Here we show that the intramolecular association with the N-terminal domain in LGN involves GL3, GL4, and a region between GL2 and GL3, whereas GL1 and GL2 do not play a major role. This conformation renders GL1 but not the other GL motifs in a state easily accessible to Gαi To interact with full-length LGN in a closed state, NuMA requires the presence of Gαi; both NuMA and Gαi are essential for cortical recruitment of LGN in mitotic cells. In contrast, mInsc, a protein that competes with NuMA for binding to LGN and regulates mitotic spindle orientation in asymmetric cell division, efficiently binds to full-length LGN without Gαi and induces its conformational change, enhancing its association with Gαi In nonpolarized symmetrically dividing HeLa cells, disruption of the LGN-NuMA interaction by ectopic expression of mInsc results in a loss of cortical localization of NuMA during metaphase and anaphase and promotes mitotic spindle misorientation and a delayed anaphase progression. These findings highlight a specific role for LGN-mediated cell cortex recruitment of NuMA.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microtúbulos/metabolismo , Huso Acromático/metabolismo , Animales , Proteínas Portadoras/metabolismo , Ciclo Celular , Perros , Células HEK293 , Células HeLa , Humanos , Células de Riñón Canino Madin Darby , Mitosis , Proteínas Nucleares/metabolismo , Dominios Proteicos
6.
Genes Cells ; 23(6): 480-493, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29718541

RESUMEN

Transmembrane glycoproteins, synthesized at the endoplasmic reticulum (ER), generally reach the Golgi apparatus in COPII-coated vesicles en route to the cell surface. Here, we show that the bona fide nonglycoprotein Nox5, a transmembrane superoxide-producing NADPH oxidase, is transported to the cell surface in a manner resistant to co-expression of Sar1 (H79G), a GTP-fixed mutant of the small GTPase Sar1, which blocks COPII vesicle fission from the ER. In contrast, Sar1 (H79G) effectively inhibits ER-to-Golgi transport of glycoproteins including the Nox5-related oxidase Nox2. The trafficking of Nox2, but not that of Nox5, is highly sensitive to over-expression of syntaxin 5 (Stx5), a t-SNARE required for COPII ER-to-Golgi transport. Thus, Nox2 and Nox5 mainly traffic via the Sar1/Stx5-dependent and -independent pathways, respectively. Both participate in Nox1 trafficking, as Nox1 advances to the cell surface in two differentially N-glycosylated forms, one complex and one high mannose, in a Sar1/Stx5-dependent and -independent manner, respectively. Nox2 and Nox5 also can use both pathways: a glycosylation-defective mutant Nox2 is weakly recruited to the plasma membrane in a less Sar1-dependent manner; N-glycosylated Nox5 mutants reach the cell surface in part as the complex form Sar1-dependently, albeit mainly as the high-mannose form in a Sar1-independent manner.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , NADPH Oxidasa 5/metabolismo , Superóxidos/metabolismo , Secuencia de Aminoácidos , Retículo Endoplásmico/metabolismo , Glicosilación , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Mutación , NADPH Oxidasa 1/genética , NADPH Oxidasa 1/metabolismo , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , Transporte de Proteínas , Homología de Secuencia
7.
Genes Cells ; 22(3): 293-309, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28185378

RESUMEN

Correct cyst morphogenesis of epithelial cells requires apical-basal polarization, which is partly regulated by mitotic spindle orientation, a process dependent on the heterotrimeric G protein subunit Gαi and its binding protein LGN. Here, we show that in three-dimensional culture of mammalian epithelial Madin-Darby canine kidney (MDCK) cells, the Gαi-activating protein Ric-8A is crucial for orientation of the mitotic spindle and formation of normal cysts that comprise a single layer of polarized cells with their apical surfaces lining an inner lumen. Consistent with the involvement of LGN, cystogenesis can be well organized by ADP-ribosylated Gαi, retaining the ability to interact with LGN, but not by the interaction-defective mutant protein Gαi2 (N150I). In monolayer culture of MDCK cells, functional tight junction (TJ) assembly, a process associated with epithelial cell polarization, is significantly delayed in Ric-8A-depleted cells as well as in Gαi-depleted cells in a mitosis-independent manner. Ric-8A knockdown results in a delayed cortical delivery of Gαi and the apical membrane protein gp135, and an increased formation of intercellular lumens surrounded by membranes rich in Gαi3 and gp135. TJ development also involves LGN and its related protein AGS3. Thus, Ric-8A regulates mammalian epithelial cell polarity for TJ assembly and cystogenesis probably in concert with Gαi and LGN/AGS3.


Asunto(s)
Células Epiteliales/fisiología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Uniones Estrechas/metabolismo , Animales , Polaridad Celular , Perros , Células de Riñón Canino Madin Darby , Mitosis , Organogénesis , Transporte de Proteínas , Huso Acromático/metabolismo , Huso Acromático/ultraestructura
8.
J Biol Chem ; 291(7): 3333-45, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26694615

RESUMEN

Inscuteable (Insc) regulates cell fate decisions in several types of stem cells. Although it is recognized that the expression levels of mouse INSC govern the balance between symmetric and asymmetric stem cell division, regulation of mouse Insc gene expression remains poorly understood. Here, we showed that mouse Insc expression transiently increases at an early stage of differentiation, when mouse embryonic stem (mES) cells differentiate into bipotent mesendoderm capable of producing both endoderm and mesoderm in defined culture conditions. We identified the minimum transcriptional regulatory element (354 bases) that drives mouse Insc transcription in mES cells within a region >5 kb upstream of the mouse Insc transcription start site. We found that the transcription factor reticuloendotheliosis oncogene (c-Rel) bound to the minimum element and promoted mouse Insc expression in mES cells. In addition, short interfering RNA-mediated knockdown of either mouse INSC or c-Rel protein decreased mesodermal cell populations without affecting differentiation into the mesendoderm or endoderm. Furthermore, overexpression of mouse INSC rescued the mesoderm-reduced phenotype induced by knockdown of c-Rel. We propose that regulation of mouse Insc expression by c-Rel modulates cell fate decisions during mES cell differentiation.


Asunto(s)
Proteínas de Ciclo Celular/agonistas , Diferenciación Celular , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Endodermo/citología , Endodermo/metabolismo , Genes Reporteros , Proteína Goosecoide/genética , Proteína Goosecoide/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Células Madre Embrionarias de Ratones/citología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-rel/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-rel/genética , Interferencia de ARN , ARN Interferente Pequeño , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Elementos Reguladores de la Transcripción , Sitio de Iniciación de la Transcripción
9.
Biochem Biophys Res Commun ; 483(3): 941-945, 2017 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-28082199

RESUMEN

The heterotrimeric G protein subunit Gαi can be activated by G protein-coupled receptors and the cytosolic protein Ric-8A, the latter of which is also known to prevent ubiquitin-dependent degradation of Gαi. Here we show that the amounts of the three Gαi-related proteins Gαi1, Gαi2, and Gαi3, but not that of Gαq, are rapidly decreased by cell treatment with pertussis toxin (PTX). The decrease appears to be due to ADP-ribosylation of Gαi, because PTX treatment does not affect the amount of a mutant Gαi2 carrying alanine substitution for Cys352, the residue that is ADP-ribosylated by the toxin. The presence of endogenous and exogenous Ric-8A increases Gαi stability as shown in cells treated with the protein synthesis inhibitor cycloheximide; however, Ric-8A fails to efficiently stabilize ADP-ribosylated Gαi. The failure agrees with the inability of Ric-8A to bind to ADP-ribosylated Gαi both in vitro and in vivo. Thus PTX appears to exert its pathological effects at least in part by converting Gαi to an unstable ADP-ribosylated form, in addition to the well-known inability of ADP-ribosylated Gαi to transduce signals triggered by G protein-coupled receptors.


Asunto(s)
Adenosina Difosfato Ribosa/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Toxina del Pertussis/toxicidad , Animales , Células COS , Chlorocebus aethiops , Perros , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Técnicas de Silenciamiento del Gen , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Células de Riñón Canino Madin Darby , Ratones , Estabilidad Proteica/efectos de los fármacos , Células RAW 264.7 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
10.
Biochem J ; 467(1): 103-13, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25602620

RESUMEN

The functions of microtubules are controlled in part by tubulin post-translational modification including acetylation of Lys4° in α-tubulin. αTAT1 (α-tubulin acetyltransferase 1), an enzyme evolutionarily conserved among eukaryotes, has recently been identified as the major α-tubulin Lys4° acetyltransferase, in which AcCoA (acetyl-CoA) serves as an acetyl group donor. The regulation and substrate recognition of this enzyme, however, have not been fully understood. In the present study, we show that AcCoA and CoA each form a stable complex with human αTAT1 to maintain the protein integrity both in vivo and in vitro. The invariant residues Arg¹³² and Ser¹6° in αTAT1 participate in the stable interaction not only with AcCoA but also with CoA, which is supported by analysis of the present crystal structures of the αTAT1 catalytic domain in complex with CoA. Alanine substitution for Arg¹³² or Ser¹6° leads to a drastic misfolding of the isolated αTAT1 catalytic domain in the absence of CoA and AcCoA but not in the presence of excess amounts of either cofactor. A mutant αTAT1 carrying the R132A or S160A substitution is degraded much faster than the wild-type protein when expressed in mammalian Madin-Darby canine kidney cells. Furthermore, alanine-scanning experiments using Lys4°-containing peptides reveal that α-tubulin Ser³8 is crucial for substrate recognition of αTAT1, whereas Asp³9, Ile4², the glycine stretch (amino acid residues 43-45) and Asp46 are also involved. The requirement for substrate selection is totally different from that in various histone acetyltransferases, which appears to be consistent with the inability of αTAT1 to acetylate histones.


Asunto(s)
Acetilcoenzima A/metabolismo , Acetiltransferasas/metabolismo , Coenzima A/metabolismo , Modelos Moleculares , Fragmentos de Péptidos/metabolismo , Tubulina (Proteína)/metabolismo , Acetilcoenzima A/química , Acetiltransferasas/química , Acetiltransferasas/genética , Sustitución de Aminoácidos , Animales , Arginina/química , Dominio Catalítico , Coenzima A/química , Cristalografía por Rayos X , Perros , Estabilidad de Enzimas , Humanos , Células de Riñón Canino Madin Darby , Conformación Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Serina/química , Especificidad por Sustrato , Tubulina (Proteína)/química , Tubulina (Proteína)/genética
11.
Biochem J ; 451(2): 195-204, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23368718

RESUMEN

Bipolar spindle assembly in mitotic cells is a prerequisite to ensure correct alignment of chromosomes for their segregation to each daughter cell; spindle microtubules are tethered at plus ends to chromosomes and focused at minus ends to either of the two spindle poles. NuMA (nuclear mitotic apparatus protein) is present solely in the nucleus in interphase cells, but relocalizes during mitosis to the spindle poles to play a crucial role in spindle assembly via focusing spindle microtubules to each pole. In the present study we show that the kinesin-5 family motor Eg5 is a protein that directly interacts with NuMA, using a proteomics approach and various binding assays both in vivo and in vitro. During mitosis Eg5 appears to interact with NuMA in the vicinity of the spindle poles, whereas the interaction does not occur in interphase cells, where Eg5 is distributed throughout the cytoplasm but NuMA exclusively localizes to the nucleus. Slight, but significant, depletion of Eg5 in HeLa cells by RNA interference results in formation of less-focused spindle poles with misaligned chromosomes in metaphase; these phenotypes are similar to those induced by depletion of NuMA. Since NuMA is less accumulated at the spindle poles in Eg5-depleted cells, Eg5 probably contributes to spindle assembly via regulating NuMA localization. Furthermore, depletion of cytoplasmic dynein induces mislocalization of NuMA and phenotypes similar to those observed in NuMA-depleted cells, without affecting Eg5 localization to the spindles. Thus dynein appears to control NuMA function in conjunction with Eg5.


Asunto(s)
Antígenos Nucleares/metabolismo , Cinesinas/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Huso Acromático/fisiología , Antígenos Nucleares/genética , Proteínas de Ciclo Celular , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Dineínas/genética , Dineínas/metabolismo , Células HeLa , Humanos , Interfase/fisiología , Cinesinas/genética , Mitosis , Proteínas Asociadas a Matriz Nuclear/genética
12.
Proc Natl Acad Sci U S A ; 108(48): 19210-5, 2011 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-22074847

RESUMEN

Interaction between the mammalian cell polarity proteins mInsc (mammalian homologue of Inscuteable) and Leu-Gly-Asn repeat-enriched protein (LGN), as well as that between their respective Drosophila homologues Inscuteable and Partner of Inscuteable (Pins), plays crucial roles in mitotic spindle orientation, a process contributing to asymmetric cell division. Here, we report a crystal structure of the LGN-binding domain (LBD) of human mInsc complexed with the N-terminal tetratricopeptide repeat (TPR) motifs of human LGN at 2.6-Å resolution. In the complex, mInsc-LBD adopts an elongated structure with three binding modules--an α-helix, an extended region, and a ß-sheet connected with a loop--that runs antiparallel to LGN along the concave surface of the superhelix formed by the TPRs. Structural analysis and structure-based mutagenesis define residues that are critical for mInsc-LGN association, and reveal that the activator of G-protein signaling 3 (AGS3)-binding protein Frmpd1 [4.1/ezrin/radixin/moesin (FERM) and PSD-95/Dlg/ZO-1 (PDZ) domain-containing protein 1] and its relative Frmpd4 interact with LGN via a region homologous to a part of mInsc-LBD, whereas nuclear mitotic apparatus protein (NuMA) and the C terminus of LGN recognize the TPR domain in a manner different from that by mInsc. mInsc binds to LGN with the highest affinity (K(D) ≈ 2.4 nM) and effectively replaces the Frmpd proteins, NuMA, and the LGN C terminus, suggesting the priority of mInsc in binding to LGN. We also demonstrate, using mutant proteins, that mInsc-LGN interaction is vital for stabilization of LGN and for intracellular localization of mInsc.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Polaridad Celular/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Conformación Proteica , Huso Acromático/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Portadoras/metabolismo , Cromatografía en Gel , Cristalización , Fluorescencia , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Mutagénesis , Resonancia por Plasmón de Superficie , Ultracentrifugación
13.
Proc Natl Acad Sci U S A ; 108(7): 2861-6, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282602

RESUMEN

Regulated synthesis of reactive oxygen species (ROS) by membrane-bound fungal NADPH oxidases (Nox) plays a key role in fungal morphogenesis, growth, and development. Generation of reactive oxygen species (ROS) by the plant symbiotic fungus, Epichloë festucae, requires functional assembly of a multisubunit complex composed of NoxA, a regulatory component, NoxR, and the small GTPase RacA. However, the mechanism for assembly and activation of this complex at the plasma membrane is unknown. We found by yeast two-hybrid and coimmunoprecipitation assays that E. festucae NoxR interacts with homologs of the yeast polarity proteins, Bem1 and Cdc24, and that the Phox and Bem1 (PB1) protein domains found in these proteins are essential for these interactions. GFP fusions of BemA, Cdc24, and NoxR preferentially localized to actively growing hyphal tips and to septa. These proteins interact with each other in vivo at these same cellular sites as shown by bimolecular fluorescent complementation assays. The PB1 domain of NoxR is essential for localization to the hyphal tip. An E. festucae ΔbemA mutant was defective in hyphal morphogenesis and growth in culture and in planta. The changes in fungal growth in planta resulted in a defective symbiotic interaction phenotype. Our inability to isolate a Δcdc24 mutant suggests this gene is essential. These results demonstrate that BemA and Cdc24 play a critical role in localizing NoxR protein to sites of fungal hyphal morphogenesis and growth. Our findings identify a potential shared ancestral link between the protein machinery required for fungal polarity establishment and the Nox complex controlling cellular differentiation.


Asunto(s)
Epichloe/genética , Proteínas Fúngicas/genética , Hifa/crecimiento & desarrollo , Hifa/metabolismo , Lolium/microbiología , Complejos Multiproteicos/genética , NADPH Oxidasas/metabolismo , Simbiosis , Secuencia de Bases , Biología Computacional , Proteínas Fúngicas/metabolismo , Proteínas Fluorescentes Verdes , Inmunoprecipitación , Microscopía Confocal , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Estructura Terciaria de Proteína/genética , Especies Reactivas de Oxígeno/metabolismo , Análisis de Secuencia de ADN , Técnicas del Sistema de Dos Híbridos
14.
Biochem Biophys Res Commun ; 435(3): 414-9, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23665327

RESUMEN

The cytosolic protein Ric-8A acts as a guanine nucleotide exchange factor for Gα subunits of the Gi, Gq, and G12/13 classes of heterotrimeric G protein in vitro, and is also known to increase the amounts of these Gα proteins in vivo. The mechanism whereby Ric-8 regulates Gα content, however, has not been fully understood. Here we show that Ric-8 Astabilizes Gαi2 and Gαq by preventing their ubiquitination. Ric-8A interacts with and stabilizes Gαi2, Gαq, Gα12, but not Gαs, when expressed in COS-7 cells. The protein levels of Gαi2 and Gαq appear to be controlled via the ubiquitin-proteasome degradation pathway, because these Gα subunits undergo polyubiquitination and are stabilized with the proteasome inhibitor MG132. The ubiquitination of Gαi2 and Gαq is suppressed by expression of Ric-8A. The suppression likely requires Ric-8A interaction with these Gα proteins; the C-terminal truncation of Gαq and Gαi2 completely abrogates their interaction with Ric-8A, their stabilization by Ric-8A, and Ric-8A-mediated inhibition of Gα ubiquitination.


Asunto(s)
Subunidad alfa de la Proteína de Unión al GTP Gi2/antagonistas & inhibidores , Subunidad alfa de la Proteína de Unión al GTP Gi2/química , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/química , Factores de Intercambio de Guanina Nucleótido/fisiología , Animales , Células COS , Chlorocebus aethiops , Subunidad alfa de la Proteína de Unión al GTP Gi2/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Estabilidad Proteica , Ubiquitinación
15.
Genes Cells ; 15(5): 409-24, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20384786

RESUMEN

Neutrophils play an essential role via phagocytosis in host defense against microbial infections. However, little is known about molecular mechanisms underlying phagocytosis in neutrophils, because of the difficulty in genetically manipulating these cells. Here, we provide the first comprehensive description of phospholipid metabolism during phagocytosis in human neutrophils, which we have efficiently transfected with cDNAs encoding lipid-probing protein modules. Phosphatidylinositol 4,5-bisphosphate (PtdIns(4,5)P(2)), an F-actin organizer abundant in the plasma membrane, diminishes progressively from phagosomes during phagosome formation and vanishes after phagosome closure with F-actin disappearance. Diacylglycerol, a metabolite of PtdIns(4,5)P(2), appears at phagocytic cups and remains associated with nascent (closed) phagosomes; it may function with phosphatidylserine, present in both plasma and phagosomal membranes, to recruit phagocytosis-associated proteins. From PtdIns(4,5)P(2), PtdIns(3,4,5)P(3) is also produced at phagocytic cups but becomes undetectable shortly after phagosome sealing, consistent with its proposed roles in pseudopod extension and phagosome closure. PtdIns(3)P, a putative participant in phagosome maturation, emerges at closed phagosomes as does the class III PtdIns 3-kinase Vps34. Although the small GTPases Rab5 and Rab7 are thought to contribute to phagosome maturation in macrophages, Rab5 but not Rab7 fails to accumulate at phagosomes in neutrophils, suggesting a difference in phagocytic mechanism between the two phagocytes.


Asunto(s)
Membrana Celular/metabolismo , Neutrófilos/metabolismo , Neutrófilos/fisiología , Fagocitosis/fisiología , Fosfolípidos/metabolismo , Actinas/metabolismo , Actinas/ultraestructura , Animales , Membrana Celular/química , Células Cultivadas , Activación Enzimática , Humanos , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Neutrófilos/citología , Fagosomas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo , Proteínas de Unión a GTP rab7
16.
Nat Cell Biol ; 6(6): 547-54, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15156153

RESUMEN

Although the Notch and JAK-STAT signalling pathways fulfill overlapping roles in growth and differentiation regulation, no coordination mechanism has been proposed to explain their relationship. Here we show that STAT3 is activated in the presence of active Notch, as well as the Notch effectors Hes1 and Hes5. Hes proteins associate with JAK2 and STAT3, and facilitate complex formation between JAK2 and STAT3, thus promoting STAT3 phosphorylation and activation. Furthermore, suppression of endogenous Hes1 expression reduces growth factor induction of STAT3 phosphorylation. STAT3 seems to be essential for maintenance of radial glial cells and differentiation of astrocytes by Notch in the developing central nervous system. These results suggest that direct protein-protein interactions coordinate cross-talk between the Notch-Hes and JAK-STAT pathways.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas , Receptores de Superficie Celular/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factores de Transcripción , Animales , Astrocitos/citología , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células COS , Diferenciación Celular/fisiología , Sistema Nervioso Central/citología , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Proteínas de Unión al ADN/genética , Feto , Janus Quinasa 2 , Ratones , Fosforilación , Receptor Notch1 , Receptores de Superficie Celular/genética , Proteínas Represoras/genética , Factor de Transcripción STAT3 , Transactivadores/genética , Factor de Transcripción HES-1
17.
Biochem J ; 419(2): 329-38, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19090790

RESUMEN

The superoxide-producing NADPH oxidase in phagocytes is crucial for host defence; its catalytic core is the membrane-integrated protein gp91phox [also known as Nox2 (NADPH oxidase 2)], which forms a stable heterodimer with p22phox. Activation of the oxidase requires membrane translocation of the three cytosolic proteins p47phox, p67phox and the small GTPase Rac. At the membrane, these proteins assemble with the gp91phox-p22phox heterodimer and induce a conformational change of gp91phox, leading to superoxide production. p47phox translocates to membranes using its two tandemly arranged SH3 domains, which directly interact with p22phox, whereas p67phox is recruited in a p47phox-dependent manner. In the present study, we show that a short region N-terminal to the bis-SH3 domain is required for activation of the phagocyte NADPH oxidase. Alanine substitution for Ile152 in this region, a residue that is completely conserved during evolution, results in a loss of the ability to activate the oxidase; and the replacement of Thr153 also prevents oxidase activation, but to a lesser extent. In addition, the corresponding isoleucine residue (Ile155) of the p47phox homologue Noxo1 (Nox organizer 1) participates in the activation of non-phagocytic oxidases, such as Nox1 and Nox3. The I152A substitution in p47phox, however, does not affect its interaction with p22phox or with p67phox. Consistent with this, a mutant p47phox (I152A), as well as the wild-type protein, is targeted upon cell stimulation to membranes, and membrane recruitment of p67phox and Rac normally occurs in p47phox (I152A)-expressing cells. Thus the Ile152-containing region of p47phox plays a crucial role in oxidase activation, probably by functioning at a process after oxidase assembly.


Asunto(s)
NADPH Oxidasas/metabolismo , Fagocitos/enzimología , Dominios Homologos src/fisiología , Animales , Transporte Biológico/genética , Transporte Biológico/fisiología , Células CHO , Células COS , Línea Celular , Membrana Celular/metabolismo , Chlorocebus aethiops , Cricetinae , Cricetulus , Humanos , Isoleucina/genética , Isoleucina/fisiología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , NADPH Oxidasa 1 , NADPH Oxidasa 2 , NADPH Oxidasas/genética , NADPH Oxidasas/fisiología , Neutrófilos/metabolismo , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfoproteínas/fisiología , Unión Proteica/genética , Unión Proteica/fisiología , Relación Estructura-Actividad , Treonina/genética , Treonina/fisiología , Proteínas de Unión al GTP rac/genética , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rac/fisiología , Dominios Homologos src/genética
18.
Sci STKE ; 2007(401): re6, 2007 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-17726178

RESUMEN

Proteins containing the PB1 domain, a protein interaction module conserved in animals, fungi, amoebas, and plants, participate in diverse biological processes. The PB1 domains adopt a ubiquitin-like beta-grasp fold, containing two alpha helices and a mixed five-stranded beta sheet, and are classified into groups harboring an acidic OPCA motif (type I), the invariant lysine residue on the first beta strand (type II), or both (type I/II). The OPCA motif of a type I PB1 domain forms salt bridges with basic residues, especially the conserved lysine, of a type II PB1 domain, thereby mediating a specific PB1-PB1 heterodimerization, whereas additional contacts contribute to high affinity and specificity of the modular interaction. The canonical PB1 dimerization is required for the formation of complexes between p40(phox) and p67(phox) (for activation of the NADPH oxidase crucial for mammalian host defense), between the scaffold Bem1 and the guanine nucleotide exchange factor Cdc24 (for polarity establishment in yeasts), and between the polarity protein Par6 and atypical protein kinase C (for cell polarization in animal cells), as well as for the interaction between the mitogen-activated protein kinase kinase kinases MEKK2 or MEKK3 and the downstream target mitogen-activated protein kinase kinase MEK5 (for early cardiovascular development in mammals). PB1 domains can also mediate interactions with other protein domains. For example, an intramolecular interaction between the PB1 and PX domains of p40(phox) regulates phagosomal targeting of the microbicidal NADPH oxidase; the PB1 domain of MEK5 is likely responsible for binding to the downstream kinase ERK5, which lacks a PB1 domain; and the scaffold protein Nbr1 associates through a PB1-containing region with titin, a sarcomere protein without a PB1 domain. This Review describes various aspects of PB1 domains at the molecular and cellular levels.


Asunto(s)
Proteínas Fúngicas/metabolismo , Proteínas de Plantas/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas de Plantas/química , Proteínas de Plantas/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Homología de Secuencia de Aminoácido
19.
Oncotarget ; 8(22): 36211-36224, 2017 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-28423707

RESUMEN

This study aims to demonstrate the clinical and biological significance of Brain derived neurotrophic factor (BDNF)/Tropomyosin-related kinase B (TrkB) signaling in gallbladder cancer (GBC) through a series of in vitro and in vivo experiments. TrkB expression was detected in 63 (91.3%) out of 69 surgically resected primary GBC specimens by immunohistochemistry. TrkB expression in the invasive front correlated with T factor (p=0.0391) and clinical staging (p=0.0391). Overall survival was lower in patients with high TrkB expression in the invasive front than in those with low TrkB expression (p=0.0363). In vitro experiment, we used five TrkB-expressing GBC cell lines with or without K-ras mutation. TrkB-mediated signaling increased proliferation and the invasiveness by inducing epithelial mesenchymal transition, and activating matrix metalloproteinases-2 (MMP-2) and MMP-9. Inhibition of TrkB-mediated signaling also decreased hypoxia-inducible factor-1α, vascular endothelial growth factor A (VEGF-A), VEGF-C, and VEGF-D expression. In vivo experiment, inhibition of TrkB-mediated signaling suppressed tumorigenicity and tumor growth in GBC. These findings demonstrate that TrkB-mediated signaling contributes to the induction of malignant phenotypes (proliferation, invasiveness, angiogenesis, lymphangiogenesis, and tumorigenesis) in GBC, and could be a promising therapeutic target regardless of K-ras mutation status.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Neoplasias de la Vesícula Biliar/genética , Glicoproteínas de Membrana/genética , Receptor trkB/genética , Anciano , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Neoplasias de la Vesícula Biliar/mortalidad , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Estadificación de Neoplasias , ARN Interferente Pequeño/genética , Receptor trkB/metabolismo , Transducción de Señal , Análisis de Supervivencia , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
J Cell Biol ; 200(5): 635-50, 2013 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-23439680

RESUMEN

Formation of apico-basal polarity in epithelial cells is crucial for both morphogenesis (e.g., cyst formation) and function (e.g., tight junction development). Atypical protein kinase C (aPKC), complexed with Par6, is considered to translocate to the apical membrane and function in epithelial cell polarization. However, the mechanism for translocation of the Par6-aPKC complex has remained largely unknown. Here, we show that the WD40 protein Morg1 (mitogen-activated protein kinase organizer 1) directly binds to Par6 and thus facilitates apical targeting of Par6-aPKC in Madin-Darby canine kidney epithelial cells. Morg1 also interacts with the apical transmembrane protein Crumbs3 to promote Par6-aPKC binding to Crumbs3, which is reinforced with the apically localized small GTPase Cdc42. Depletion of Morg1 disrupted both tight junction development in monolayer culture and cyst formation in three-dimensional culture; apico-basal polarity was notably restored by forced targeting of aPKC to the apical surface. Thus, Par6-aPKC recruitment to the premature apical membrane appears to be required for definition of apical identity of epithelial cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Membrana Celular/enzimología , Polaridad Celular , Células Epiteliales/enzimología , Riñón/enzimología , Glicoproteínas de Membrana/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Perros , Riñón/embriología , Células de Riñón Canino Madin Darby , Glicoproteínas de Membrana/genética , Morfogénesis , Mutagénesis Sitio-Dirigida , Mutación , Unión Proteica , Proteína Quinasa C/genética , Transporte de Proteínas , Interferencia de ARN , Uniones Estrechas/enzimología , Factores de Tiempo , Transfección , Proteína de Unión al GTP cdc42/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA