Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Cancer ; 23(1): 56, 2024 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-38491381

RESUMEN

One of the major hurdles that has hindered the success of chimeric antigen receptor (CAR) T cell therapies against solid tumors is on-target off-tumor (OTOT) toxicity due to sharing of the same epitopes on normal tissues. To elevate the safety profile of CAR-T cells, an affinity/avidity fine-tuned CAR was designed enabling CAR-T cell activation only in the presence of a highly expressed tumor associated antigen (TAA) but not when recognizing the same antigen at a physiological level on healthy cells. Using direct stochastic optical reconstruction microscopy (dSTORM) which provides single-molecule resolution, and flow cytometry, we identified high carbonic anhydrase IX (CAIX) density on clear cell renal cell carcinoma (ccRCC) patient samples and low-density expression on healthy bile duct tissues. A Tet-On doxycycline-inducible CAIX expressing cell line was established to mimic various CAIX densities, providing coverage from CAIX-high skrc-59 tumor cells to CAIX-low MMNK-1 cholangiocytes. Assessing the killing of CAR-T cells, we demonstrated that low-affinity/high-avidity fine-tuned G9 CAR-T has a wider therapeutic window compared to high-affinity/high-avidity G250 that was used in the first anti-CAIX CAR-T clinical trial but displayed serious OTOT effects. To assess the therapeutic effect of G9 on patient samples, we generated ccRCC patient derived organotypic tumor spheroid (PDOTS) ex vivo cultures and demonstrated that G9 CAR-T cells exhibited superior efficacy, migration and cytokine release in these miniature tumors. Moreover, in an RCC orthotopic mouse model, G9 CAR-T cells showed enhanced tumor control compared to G250. In summary, G9 has successfully mitigated OTOT side effects and in doing so has made CAIX a druggable immunotherapeutic target.


Asunto(s)
Anhidrasas Carbónicas , Carcinoma de Células Renales , Neoplasias Renales , Receptores Quiméricos de Antígenos , Animales , Ratones , Humanos , Anhidrasa Carbónica IX/genética , Carcinoma de Células Renales/metabolismo , Neoplasias Renales/patología , Receptores Quiméricos de Antígenos/genética , Anhidrasas Carbónicas/metabolismo , Anhidrasas Carbónicas/uso terapéutico , Antígenos de Neoplasias , Anticuerpos , Linfocitos T/metabolismo
2.
J Cell Biol ; 219(7)2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32356865

RESUMEN

The Aurora B chromosomal passenger complex (CPC) is a conserved regulator of mitosis. Its functions require localization first to the chromosome arms and then centromeres in mitosis and subsequently the central spindle in anaphase. Here, we analyze the requirements for core CPC subunits, survivin and INCENP, and the mitotic kinesin-like protein 2 (MKLP2) in targeting to these distinct localizations. Centromere recruitment of the CPC requires interaction of survivin with histone H3 phosphorylated at threonine 3, and we provide a complete structure of this assembly. Furthermore, we show that the INCENP RRKKRR-motif is required for both centromeric localization of the CPC in metaphase and MKLP2-dependent transport in anaphase. MKLP2 and DNA bind competitively to this motif, and INCENP T59 phosphorylation acts as a switch preventing MKLP2 binding in metaphase. In anaphase, CPC binding promotes the microtubule-dependent ATPase activity of MKLP2. These results explain how centromere targeting of the CPC in mitosis is coupled to its movement to the central spindle in anaphase.


Asunto(s)
Anafase , Aurora Quinasa B/metabolismo , Cromatina/metabolismo , Histonas/metabolismo , Cinesinas/metabolismo , Procesamiento Proteico-Postraduccional , Secuencia de Aminoácidos , Animales , Aurora Quinasa B/química , Aurora Quinasa B/genética , Unión Competitiva , Centrómero/metabolismo , Centrómero/ultraestructura , Cromatina/ultraestructura , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , Células HeLa , Histonas/química , Histonas/genética , Humanos , Cinesinas/química , Cinesinas/genética , Metafase , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Modelos Moleculares , Fosforilación , Unión Proteica , Estructura Secundaria de Proteína , Transporte de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Huso Acromático/metabolismo , Huso Acromático/ultraestructura , Survivin/química , Survivin/genética , Survivin/metabolismo
3.
J Cell Biol ; 216(10): 3029-3039, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28860275

RESUMEN

The meiotic spindle is formed without centrosomes in a large volume of oocytes. Local activation of crucial spindle proteins around chromosomes is important for formation and maintenance of a bipolar spindle in oocytes. We found that phosphodocking 14-3-3 proteins stabilize spindle bipolarity in Drosophila melanogaster oocytes. A critical 14-3-3 target is the minus end-directed motor Ncd (human HSET; kinesin-14), which has well-documented roles in stabilizing a bipolar spindle in oocytes. Phospho docking by 14-3-3 inhibits the microtubule binding activity of the nonmotor Ncd tail. Further phosphorylation by Aurora B kinase can release Ncd from this inhibitory effect of 14-3-3. As Aurora B localizes to chromosomes and spindles, 14-3-3 facilitates specific association of Ncd with spindle microtubules by preventing Ncd from binding to nonspindle microtubules in oocytes. Therefore, 14-3-3 translates a spatial cue provided by Aurora B to target Ncd selectively to the spindle within the large volume of oocytes.


Asunto(s)
Proteínas 14-3-3/metabolismo , Cromosomas de Insectos/metabolismo , Proteínas de Drosophila/metabolismo , Cinesinas/metabolismo , Microtúbulos/metabolismo , Oocitos/metabolismo , Huso Acromático/metabolismo , Proteínas 14-3-3/genética , Animales , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Cromosomas de Insectos/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Femenino , Cinesinas/genética , Microtúbulos/genética , Oocitos/citología , Transporte de Proteínas/fisiología , Huso Acromático/genética
4.
J Cell Biol ; 207(6): 683-93, 2014 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-25512391

RESUMEN

The mitotic kinase Aurora B is concentrated at the anaphase central spindle by the kinesin MKlp2 during mitotic exit and cytokinesis. This pool of Aurora B phosphorylates substrates including the kinesin KIF4A to regulate central spindle length. In this paper, we identify a counteracting system in which PP2A-B56γ and -ε, but not PP2A-B56α, -ß, and -δ, are maintained at the central spindle by KIF4A. Biochemical assays show that PP2A-B56γ can dephosphorylate the T799 Aurora B site on KIF4A and thereby counteract the Aurora B- and microtubule-stimulated ATPase activity of KIF4A. In agreement with these observations, combined silencing of PP2A-B56γ and -ε resulted in increased phosphorylation of KIF4A T799 and decreased central spindle growth in anaphase B. Furthermore, reduced turnover of regulatory phosphorylation on another Aurora B substrate MKlp1 was observed, suggesting that PP2A-B56γ and -ε play a general role opposing Aurora B at the central spindle. KIF4A and PP2A-B56γ and -ε therefore create a spatially restricted negative feedback loop counteracting Aurora B in anaphase.


Asunto(s)
Anafase , Aurora Quinasa B/metabolismo , Cinesinas/metabolismo , Proteína Fosfatasa 2/metabolismo , Animales , Retroalimentación Fisiológica , Células HeLa , Humanos , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Células Sf9 , Huso Acromático/enzimología , Spodoptera , Imagen de Lapso de Tiempo
5.
J Cell Biol ; 206(7): 833-42, 2014 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-25246613

RESUMEN

The spindle assembly checkpoint (SAC) monitors correct attachment of chromosomes to microtubules, an important safeguard mechanism ensuring faithful chromosome segregation in eukaryotic cells. How the SAC signal is turned off once all the chromosomes have successfully attached to the spindle remains an unresolved question. Mps1 phosphorylation of Knl1 results in recruitment of the SAC proteins Bub1, Bub3, and BubR1 to the kinetochore and production of the wait-anaphase signal. SAC silencing is therefore expected to involve a phosphatase opposing Mps1. Here we demonstrate in vivo and in vitro that BubR1-associated PP2A-B56 is a key phosphatase for the removal of the Mps1-mediated Knl1 phosphorylations necessary for Bub1/BubR1 recruitment in mammalian cells. SAC silencing is thus promoted by a negative feedback loop involving the Mps1-dependent recruitment of a phosphatase opposing Mps1. Our findings extend the previously reported role for BubR1-associated PP2A-B56 in opposing Aurora B and suggest that BubR1-bound PP2A-B56 integrates kinetochore surveillance and silencing of the SAC.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Puntos de Control de la Fase M del Ciclo Celular , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Fosfatasa 2/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Células HeLa , Humanos , Cinetocoros/enzimología , Fosforilación , Transporte de Proteínas
6.
J Cell Biol ; 205(5): 707-20, 2014 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-24891604

RESUMEN

The ancestral Rab GTPase Rab18 and both subunits of the Rab3GAP complex are mutated in the human neurological and developmental disorder Warburg Micro syndrome. Here, we demonstrate that the Rab3GAP complex is a specific Rab18 guanine nucleotide exchange factor (GEF). The Rab3GAP complex localizes to the endoplasmic reticulum (ER) and is necessary for ER targeting of Rab18. It is also sufficient to promote membrane recruitment of Rab18. Disease-associated point mutations of conserved residues in either the Rab3GAP1 (T18P and E24V) or Rab3GAP2 (R426C) subunits result in loss of the Rab18 GEF and membrane-targeting activities. Supporting the view that Rab18 activity is important for ER structure, in the absence of either Rab3GAP subunit or Rab18 function, ER tubular networks marked by reticulon 4 were disrupted, and ER sheets defined by CLIMP-63 spread out into the cell periphery. Micro syndrome is therefore a disease characterized by direct loss of Rab18 function or loss of Rab18 activation at the ER by its GEF Rab3GAP.


Asunto(s)
Retículo Endoplásmico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP rab/fisiología , Animales , Células COS , Membrana Celular/metabolismo , Chlorocebus aethiops , GTP Fosfohidrolasas/metabolismo , Células HEK293 , Células HeLa , Humanos , Lípidos/química , Proteínas de la Membrana/metabolismo , Mutación , Fenotipo , Mutación Puntual , Proteínas de Unión al GTP rab3/metabolismo
7.
Mol Cell ; 52(3): 393-405, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24120663

RESUMEN

Cytokinesis follows separase activation and chromosome segregation. This order is ensured in budding yeast by the mitotic exit network (MEN), where Cdc14p dephosphorylates key conserved Cdk1-substrates exemplified by the anaphase spindle-elongation protein Ase1p. However, in metazoans, MEN and Cdc14 function is not conserved. Instead, the PP2A-B55α/ENSA/Greatwall (BEG) pathway controls the human Ase1p ortholog PRC1. In this pathway, PP2A-B55 inhibition is coupled to Cdk1-cyclin B activity, whereas separase inhibition is maintained by cyclin B concentration. This creates two cyclin B thresholds during mitotic exit. Simulation and experiments using PRC1 as a model substrate show that the first threshold permits separase activation and chromosome segregation, and the second permits PP2A-B55 activation and initiation of cytokinesis. Removal of the ENSA/Greatwall (EG) timer module eliminates this second threshold, as well as associated delay in PRC1 dephosphorylation and initiation of cytokinesis, by uncoupling PP2A-B55 from Cdk1-cyclin B activity. Therefore, temporal order during mitotic exit is promoted by the metazoan BEG pathway.


Asunto(s)
Segregación Cromosómica/genética , Citocinesis/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Fosfatasa 2/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Quinasa CDC2/metabolismo , Cromosomas/genética , Ciclina B/metabolismo , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Mitosis/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteína Fosfatasa 2/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Fosfatasas , Separasa/genética , Separasa/metabolismo , Transducción de Señal/genética
8.
J Cell Sci ; 126(Pt 15): 3429-40, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23729733

RESUMEN

Mutations in the PPP6C catalytic subunit of protein phosphatase 6 (PP6) are drivers for the development of melanoma. Here, we analyse a panel of melanoma-associated mutations in PPP6C and find that these generally compromise assembly of the PP6 holoenzyme and catalytic activity towards a model substrate. Detailed analysis of one mutant, PPP6C-H114Y, in both primary melanoma and engineered cell lines reveals it is destabilized and undergoes increased proteasome-mediated turnover. Global analysis of phosphatase substrates by mass spectrometry identifies the oncogenic kinase Aurora-A as the major PP6 substrate that is dysregulated under these conditions. Accordingly, cells lacking PPP6C or carrying the PPP6C-H114Y allele have elevated Aurora-A kinase activity and display chromosome instability with associated Aurora-A-dependent micronucleation. Chromosomes mis-segregated to these micronuclei are preferentially stained by the DNA damage marker γ-H2AX, suggesting that loss of PPP6C promotes both chromosome instability and DNA damage. These findings support the view that formation of micronuclei rather than chromosome instability alone explains how loss of PPP6C, and more generally mitotic spindle and centrosome defects, can act as drivers for genome instability in melanoma and other cancers.


Asunto(s)
Aurora Quinasa A/metabolismo , Inestabilidad Cromosómica , Daño del ADN , Melanoma/genética , Fosfoproteínas Fosfatasas/genética , Secuencia de Aminoácidos , Aurora Quinasa A/genética , Línea Celular Tumoral , Células HeLa , Humanos , Melanoma/enzimología , Melanoma/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación
9.
J Cell Biol ; 198(5): 865-80, 2012 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-22945935

RESUMEN

In mitosis, animal cells lose their adhesion to the surrounding surfaces and become rounded. During mitotic exit, they reestablish these adhesions and at the same time physically contract and divide. How these competing processes are spatially segregated at the cell cortex remains mysterious. To address this question, we define the specific effector pathways used by RhoA and Rac1 in mitotic cells. We demonstrate that the MKlp1-CYK4 centralspindlin complex is a guanosine triphosphatase-activating protein (GAP) for Rac1 and not RhoA and that CYK4 negatively regulated Rac1 activity at the cell equator in anaphase. Cells expressing a CYK4 GAP mutant had defects in cytokinesis and showed elevated staining for the cell adhesion marker vinculin. These defects could be rescued by depletion of ARHGEF7 and p21-activated kinase, Rac1-specific effector proteins required for cell adhesion. Based on these findings, we propose that CYK4 GAP activity is required during anaphase to inhibit Rac1-dependent effector pathways associated with control of cell spreading and adhesion.


Asunto(s)
Citocinesis/fisiología , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Anafase/fisiología , Adhesión Celular/fisiología , Línea Celular Tumoral , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/fisiología , Factores de Intercambio de Guanina Nucleótido Rho , Transducción de Señal , Vinculina/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
10.
J Cell Biol ; 191(7): 1315-32, 2010 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-21187329

RESUMEN

Many protein kinases are activated by a conserved regulatory step involving T-loop phosphorylation. Although there is considerable focus on kinase activator proteins, the importance of specific T-loop phosphatases reversing kinase activation has been underappreciated. We find that the protein phosphatase 6 (PP6) holoenzyme is the major T-loop phosphatase for Aurora A, an essential mitotic kinase. Loss of PP6 function by depletion of catalytic or regulatory subunits interferes with spindle formation and chromosome alignment because of increased Aurora A activity. Aurora A T-loop phosphorylation and the stability of the Aurora A-TPX2 complex are increased in cells depleted of PP6 but not other phosphatases. Furthermore, purified PP6 acts as a T-loop phosphatase for Aurora A-TPX2 complexes in vitro, whereas catalytically inactive mutants cannot dephosphorylate Aurora A or rescue the PPP6C depletion phenotype. These results demonstrate a hitherto unappreciated role for PP6 as the T-loop phosphatase regulating Aurora A activity during spindle formation and suggest the general importance of this form of regulation.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis/fisiología , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Huso Acromático/fisiología , Anafase/genética , Antígenos Nucleares/metabolismo , Aurora Quinasas , Azepinas/farmacología , Dominio Catalítico/genética , Proteínas de Ciclo Celular/genética , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/patología , Segregación Cromosómica/genética , Ciclina B/metabolismo , Fibroblastos/patología , Células HeLa , Histonas/genética , Histonas/metabolismo , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Cinesinas/antagonistas & inhibidores , Cinesinas/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Modelos Biológicos , Mutación/genética , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas Nucleares/genética , Fosfoproteínas Fosfatasas/genética , Monoéster Fosfórico Hidrolasas/genética , Fosforilación/efectos de los fármacos , Unión Proteica/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , Huso Acromático/efectos de los fármacos , Telofase/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
11.
J Cell Biol ; 191(4): 751-60, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21079244

RESUMEN

Cytokinesis requires a membrane-remodeling and fission event termed abscission that occurs after chromosome segregation, cleavage furrow formation, and contraction have completed. In this study, we show how abscission factor recruitment is controlled by the Polo-like kinase 1 (Plk1). At the metaphase-anaphase transition, Plk1 initiates cleavage furrow formation and is then progressively degraded during mitotic exit. During this period, Plk1 phosphorylates the abscission factor Cep55 in trans and prevents its untimely recruitment to the anaphase spindle. A Plk1 phosphorylation site mutant of Cep55 is prematurely recruited to the anaphase spindle and fails to support abscission. Endogenous Cep55 behaves similarly after Plk1 inhibition by the drugs BI2536 or GW842862. Only once Plk1 is degraded can Cep55 target to the midbody and promote abscission. Blocking Plk1 degradation leads to elevated levels of Plk1 at the midbody and the failure of Cep55 recruitment. Thus, Plk1 activity negatively regulates Cep55 to ensure orderly abscission factor recruitment and ensures that this occurs only once cell contraction has completed.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Citocinesis/fisiología , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Aurora Quinasas , Proteínas de Ciclo Celular/genética , Células HeLa , Humanos , Proteínas Nucleares/genética , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Huso Acromático/metabolismo
12.
Methods Mol Biol ; 313: 333-43, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16118442

RESUMEN

Proper folding, and consequently exit from the endoplasmic reticulum (ER) and secretion of heterologous exocytic proteins in yeast can be rescued by fusing the proteins to certain yeast-derived polypeptides. Biologically active mammalian glycoproteins can be produced in Saccharomyces cerevisiae and Pichia pastoris by joining them to a fragment of a natural secretory glycoprotein of S. cerevisiae, Hsp150delta. The performance of the Hsp150delta carrier in both yeasts appears to exceed that of the MFalpha leader, which is widely used in industrial protein production. Here we describe the use of the Hsp150delta carrier in P. pastoris in both shake flask and fermentor cultivations. As a reporter protein we use the periplasmic disulfide-bonded Escherichia coli enzyme beta-lactamase.


Asunto(s)
Glicoproteínas/biosíntesis , Glicoproteínas/genética , Proteínas de Choque Térmico/biosíntesis , Proteínas de Choque Térmico/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Proteínas de Saccharomyces cerevisiae/biosíntesis , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Secuencia de Bases , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Clonación Molecular , ADN Recombinante/genética , Escherichia coli/enzimología , Escherichia coli/genética , Fermentación , Genes Reporteros , Vectores Genéticos , Micología/métodos , Pichia/genética , Pichia/metabolismo , Plásmidos/genética , Transformación Genética , beta-Lactamasas/biosíntesis , beta-Lactamasas/genética
13.
Traffic ; 6(7): 562-74, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15941408

RESUMEN

Glycoproteins exit the endoplasmic reticulum (ER) of the yeast Saccharomyces cerevisiae in coat protein complex II (COPII) coated vesicles. The coat consists of the essential proteins Sec23p, Sec24p, Sec13p, Sec31p, Sar1p and Sec16p. Sec24p and its two nonessential homologues Sfb2p and Sfb3p have been suggested to serve in cargo selection. Using temperature-sensitive sec24-1 mutants, we showed previously that a secretory glycoprotein, Hsp150, does not require functional Sec24p for ER exit. Deletion of SFB2, SFB3 or both from wild type or the deletion of SFB2 from sec24-1 cells did not affect Hsp150 transport. SFB3 deletion has been reported to be lethal in sec24-1. However, here we constructed a sec24-1 Deltasfb3 and a sec24-1 Deltasfb2 Deltasfb3 strain and show that Hsp150 was secreted slowly in both. Turning off the SEC24 gene did not inhibit Hsp150 secretion either, and the lack of SEC24 expression in a Deltasfb2 Deltasfb3 deletant still allowed some secretion. The sec24-1 Deltasfb2 Deltasfb3 mutant grew slower than sec24-1. The cells were irregularly shaped, budded from random sites and contained proliferated ER at permissive temperature. At restrictive temperature, the ER formed carmellae-like proliferations. Our data indicate that ER exit may occur in vesicles lacking a full complement of Sec23p/24p and Sec13p/31p, demonstrating diversity in the composition of the COPII coat.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glicoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Secuencia de Bases , Cartilla de ADN , Eliminación de Gen , Proteínas de la Membrana/genética , Microscopía Electrónica , Transporte de Proteínas , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...