Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Immunol ; 14(3): 298-305, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23354484

RESUMEN

The role of autophagy in plasma cells is unknown. Here we found notable autophagic activity in both differentiating and long-lived plasma cells and investigated its function through the use of mice with conditional deficiency in the essential autophagic molecule Atg5 in B cells. Atg5(-/-) differentiating plasma cells had a larger endoplasmic reticulum (ER) and more ER stress signaling than did their wild-type counterparts, which led to higher expression of the transcriptional repressor Blimp-1 and immunoglobulins and more antibody secretion. The enhanced immunoglobulin synthesis was associated with less intracellular ATP and more death of mutant plasma cells, which identified an unsuspected autophagy-dependent cytoprotective trade-off between immunoglobulin synthesis and viability. In vivo, mice with conditional deficiency in Atg5 in B cells had defective antibody responses, complete selection in the bone marrow for plasma cells that escaped Atg5 deletion and fewer antigen-specific long-lived bone marrow plasma cells than did wild-type mice, despite having normal germinal center responses. Thus, autophagy is specifically required for plasma cell homeostasis and long-lived humoral immunity.


Asunto(s)
Autofagia , Linfocitos B/metabolismo , Inmunoglobulinas/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Células Plasmáticas/inmunología , Adenosina Trifosfato , Animales , Formación de Anticuerpos , Proteína 5 Relacionada con la Autofagia , Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Diferenciación Celular , Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/genética , Centro Germinal/inmunología , Homeostasis , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción/biosíntesis
2.
Mol Cell ; 50(6): 783-92, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23685074

RESUMEN

To warrant the quality of the secretory proteome, stringent control systems operate at the endoplasmic reticulum (ER)-Golgi interface, preventing the release of nonnative products. Incompletely assembled oligomeric proteins that are deemed correctly folded must rely on additional quality control mechanisms dedicated to proper assembly. Here we unveil how ERp44 cycles between cisGolgi and ER in a pH-regulated manner, patrolling assembly of disulfide-linked oligomers such as IgM and adiponectin. At neutral, ER-equivalent pH, the ERp44 carboxy-terminal tail occludes the substrate-binding site. At the lower pH of the cisGolgi, conformational rearrangements of this peptide, likely involving protonation of ERp44's active cysteine, simultaneously unmask the substrate binding site and -RDEL motif, allowing capture of orphan secretory protein subunits and ER retrieval via KDEL receptors. The ERp44 assembly control cycle couples secretion fidelity and efficiency downstream of the calnexin/calreticulin and BiP-dependent quality control cycles.


Asunto(s)
Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Multimerización de Proteína , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Ciclo Celular , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis Sitio-Dirigida , Oxidorreductasas/metabolismo , Transporte de Proteínas , Vías Secretoras
3.
Proc Natl Acad Sci U S A ; 114(41): E8575-E8584, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28973899

RESUMEN

Professional secretory cells can produce large amounts of high-quality complex molecules, including IgM antibodies. Owing to their multivalency, polymeric IgM antibodies provide an efficient first-line of defense against pathogens. To decipher the mechanisms of IgM assembly, we investigated its biosynthesis in living cells and faithfully reconstituted the underlying processes in vitro. We find that a conserved peptide extension at the C-terminal end of the IgM heavy (Ig-µ) chains, termed the tailpiece, is necessary and sufficient to establish the correct geometry. Alanine scanning revealed that hydrophobic amino acids in the first half of the tailpiece contain essential information for generating the correct topology. Assembly is triggered by the formation of a disulfide bond linking two tailpieces. This induces conformational changes in the tailpiece and the adjacent domain, which drive further polymerization. Thus, the biogenesis of large and topologically challenging IgM complexes is dictated by a local conformational switch in a peptide extension.


Asunto(s)
Inmunoglobulina M/metabolismo , Cadenas mu de Inmunoglobulina/metabolismo , Fragmentos de Péptidos/metabolismo , Células HEK293 , Humanos , Inmunoglobulina M/química , Cadenas mu de Inmunoglobulina/química , Fragmentos de Péptidos/química , Multimerización de Proteína
4.
J Cell Sci ; 127(Pt 19): 4260-9, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25097228

RESUMEN

ERp44 is a pH-regulated chaperone of the secretory pathway. In the acidic milieu of the Golgi, its C-terminal tail changes conformation, simultaneously exposing the substrate-binding site for cargo capture and the RDEL motif for ER retrieval through interactions with cognate receptors. Protonation of cysteine 29 in the active site allows tail movements in vitro and in vivo. Here, we show that conserved histidine residues in the C-terminal tail also regulate ERp44 in vivo. Mutants lacking these histidine residues retain substrates more efficiently. Surprisingly, they are also O-glycosylated and partially secreted. Co-expression of client proteins prevents secretion of the histidine mutants, forcing tail opening and RDEL accessibility. Client-induced RDEL exposure allows retrieval of proteins from distinct stations along the secretory pathway, as indicated by the changes in O-glycosylation patterns upon overexpression of different partners. The ensuing gradients might help to optimize folding and assembly of different cargoes. Endogenous ERp44 is O-glycosylated and secreted by human primary endometrial cells, suggesting possible pathophysiological roles of these processes.


Asunto(s)
Proteínas Portadoras/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Humanos , Chaperonas Moleculares/genética , Control de Calidad , Vías Secretoras
5.
J Biol Chem ; 288(41): 29586-94, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23979138

RESUMEN

In the early secretory compartment (ESC), a network of chaperones and enzymes assists oxidative folding of nascent proteins. Ero1 flavoproteins oxidize protein disulfide isomerase (PDI), generating H2O2 as a byproduct. Peroxiredoxin 4 (Prx4) can utilize luminal H2O2 to oxidize PDI, thus favoring oxidative folding while limiting oxidative stress. Interestingly, neither ER oxidase contains known ER retention signal(s), raising the question of how cells prevent their secretion. Here we show that the two proteins share similar intracellular localization mechanisms. Their secretion is prevented by sequential interactions with PDI and ERp44, two resident proteins of the ESC-bearing KDEL-like motifs. PDI binds preferentially Ero1α, whereas ERp44 equally retains Ero1α and Prx4. The different binding properties of Ero1α and Prx4 increase the robustness of ER redox homeostasis.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Oxidorreductasas/metabolismo , Peroxirredoxinas/metabolismo , Vías Secretoras , Secuencia de Aminoácidos , Western Blotting , Retículo Endoplásmico/metabolismo , Células HEK293 , Células HeLa , Homeostasis , Humanos , Cinética , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Confocal , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Mutación , Oxidación-Reducción , Oxidorreductasas/genética , Peroxirredoxinas/genética , Unión Proteica , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Interferencia de ARN , Resonancia por Plasmón de Superficie
6.
Biochem Cell Biol ; 92(2): 113-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24697695

RESUMEN

To maintain proteostasis in the endoplasmic reticulum (ER), terminally misfolded secretory proteins must be recognized, partially unfolded, and dislocated to the cytosol for proteasomal destruction, in a complex process called ER-associated degradation (ERAD). Dislocation implies reduction of inter-chain disulphide bonds. When in its reduced form, protein disulphide isomerase (PDI) can act not only as a reductase but also as an unfoldase, preparing substrates for dislocation. PDI oxidation by Ero1 favours substrate release and transport across the ER membrane. Here we addressed the redox dependency of ERAD and found that DTT stimulates the dislocation of proteins with DTT-resistant disulphide bonds (i.e., orphan Ig-µ chains) but stabilizes a ribophorin mutant (Ri332) devoid of them. DTT promotes the association of Ri332, but not of Ig-µ, with PDI. This discrepancy may suggest that disulphide bonds in cargo proteins can be utilized to oxidize PDI, hence facilitating substrate detachment and degradation also in the absence of Ero1. Accordingly, Ero1 silencing retards Ri332 degradation, but has little if any effect on Ig-µ. Thus, some disulphides can increase the stability and simultaneously favour quality control of secretory proteins.


Asunto(s)
Disulfuros/metabolismo , Degradación Asociada con el Retículo Endoplásmico , Proteínas/metabolismo , Animales , Ditiotreitol/farmacología , Células HEK293 , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Proteína Disulfuro Isomerasas/metabolismo
7.
Front Cell Neurosci ; 17: 1231493, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37964795

RESUMEN

CDKL5 is a kinase with relevant functions in correct neuronal development and in the shaping of synapses. A decrease in its expression or activity leads to a severe neurodevelopmental condition known as CDKL5 deficiency disorder (CDD). CDD arises from CDKL5 mutations that lie in the coding region of the gene. However, the identification of a SNP in the CDKL5 5'UTR in a patient with symptoms consistent with CDD, together with the complexity of the CDKL5 transcript leader, points toward a relevant translational regulation of CDKL5 expression with important consequences in physiological processes as well as in the pathogenesis of CDD. We performed a bioinformatics and molecular analysis of the 5'UTR of CDKL5 to identify translational regulatory features. We propose an important role for structural cis-acting elements, with the involvement of the eukaryotic translational initiation factor eIF4B. By evaluating both cap-dependent and cap-independent translation initiation, we suggest the presence of an IRES supporting the translation of CDKL5 mRNA and propose a pathogenic effect of the C>T -189 SNP in decreasing the translation of the downstream protein.

8.
Traffic ; 11(7): 947-57, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20406418

RESUMEN

In many protein storage diseases, detergent-insoluble proteins accumulate in the early secretory compartment (ESC). Protein condensation reflects imbalances between entry into (synthesis/translocation) and exit from (secretion/degradation) ESC, and can be also a consequence of altered quality control (QC) mechanisms. Here we exploit the inducible formation of Russell bodies (RB), dilated ESC cisternae containing mutant Ig-micro chains, as a model to mechanistically dissect protein condensation. Depending on the presence or absence of Ig-L chains, mutant Ig-micro chains lacking their first constant domain (Ch1) accumulate in rough or smooth RB (rRB and sRB), dilations of the endoplasmic reticulum (ER) and ER-Golgi intermediate compartment (ERGIC), respectively, reflecting the proximal and distal QC stations in the stepwise biogenesis of polymeric IgM. Either weakening ERp44-dependent distal QC or facilitating ER-associated degradation (ERAD) inhibits RB formation. Overexpression of PDI or ERp44 inhibits muDeltaCh1 secretion. However, PDI inhibits while ERp44 promotes muDeltaCh1 condensation. Both Ero1alpha silencing and overexpression prevent RB formation, demonstrating a strict redox dependency of the phenomenon. Altogether, our findings identify key controllers of protein condensation along the ESC as potential targets to handle certain storage disorders.


Asunto(s)
Retículo Endoplásmico/metabolismo , Animales , Retículo Endoplásmico/genética , Retículo Endoplásmico/patología , Células HeLa , Humanos , Inmunoglobulina M/genética , Inmunoglobulina M/metabolismo , Polímeros/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas/genética , Proteínas/genética , Proteínas/metabolismo , Control de Calidad
9.
J Biol Chem ; 286(18): 16261-71, 2011 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-21398518

RESUMEN

In the endoplasmic reticulum (ER) of human cells, ERO1α and protein-disulfide isomerase (PDI) constitute one of the major electron flow pathways that catalyze oxidative folding of secretory proteins. Specific and limited PDI oxidation by ERO1α is essential to avoid ER hyperoxidation. To investigate how ERO1α oxidizes PDI selectively among more than 20 ER-resident PDI family member proteins, we performed docking simulations and systematic biochemical analyses. Our findings reveal that a protruding ß-hairpin of ERO1α specifically interacts with the hydrophobic pocket present in the redox-inactive PDI b'-domain through the stacks between their aromatic residues, leading to preferred oxidation of the C-terminal PDI a'-domain. ERO1α associated preferentially with reduced PDI, explaining the stepwise disulfide shuttle mechanism, first from ERO1α to PDI and then from oxidized PDI to an unfolded polypeptide bound to its hydrophobic pocket. The interaction of ERO1α with ERp44, another PDI family member protein, was also analyzed. Notably, ERO1α-dependent PDI oxidation was inhibited by a hyperactive ERp44 mutant that lacks the C-terminal tail concealing the substrate-binding hydrophobic regions. The potential ability of ERp44 to inhibit ERO1α activity may suggest its physiological role in ER redox and protein homeostasis.


Asunto(s)
Disulfuros/química , Glicoproteínas de Membrana/química , Oxidorreductasas/química , Proteína Disulfuro Isomerasas/química , Pliegue de Proteína , Sitios de Unión , Catálisis , Disulfuros/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/metabolismo , Estructura Secundaria de Proteína
10.
Elife ; 112022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36281643

RESUMEN

Hepatic metastases are a poor prognostic factor of colorectal carcinoma (CRC) and new strategies to reduce the risk of liver CRC colonization are highly needed. Herein, we used mouse models of hepatic metastatization to demonstrate that the continuous infusion of therapeutic doses of interferon-alpha (IFNα) controls CRC invasion by acting on hepatic endothelial cells (HECs). Mechanistically, IFNα promoted the development of a vascular antimetastatic niche characterized by liver sinusoidal endothelial cells (LSECs) defenestration extracellular matrix and glycocalyx deposition, thus strengthening the liver vascular barrier impairing CRC trans-sinusoidal migration, without requiring a direct action on tumor cells, hepatic stellate cells, hepatocytes, or liver dendritic cells (DCs), Kupffer cells (KCs) and liver capsular macrophages (LCMs). Moreover, IFNα endowed LSECs with efficient cross-priming potential that, along with the early intravascular tumor burden reduction, supported the generation of antitumor CD8+ T cells and ultimately led to the establishment of a protective long-term memory T cell response. These findings provide a rationale for the use of continuous IFNα therapy in perioperative settings to reduce CRC metastatic spreading to the liver.


Colorectal cancer remains one of the most widespread and deadly cancers worldwide. Poor health outcomes are usually linked to diseased cells spreading from the intestine to create new tumors in the liver or other parts of the body. Treatment involves surgically removing the initial tumors in the bowel, but patient survival could be improved if, in parallel, their immune system was 'boosted' to destroy cancer cells before they can form other tumors. Interferon alpha is a small protein which helps to coordinate how the immune system recognizes and deactivates foreign agents and cancerous cells. It has recently been trialed as a colorectal cancer treatment to prevent tumors from spreading to the liver, but only with limited success. This partly because interferon-alpha is usually administered in high and pulsed doses, which cause severe side effects through the body. Instead, Tran, Ferreira, Alvarez-Moya et al. aimed to investigate whether continuously delivering lower amounts of the drug could be a better approach. This strategy was tested on mice in which colorectal cancer cells had been implanted into the wall of the large intestine. Continuous administration minimized the risk of the implanted cancer cells spreading to the liver while also creating fewer side effects. The team was able to identify an optimum delivery strategy by varying how much interferon-alpha the animals received and when. Further experiments also revealed a new mechanism by which interferon-alpha prevented the spread of colorectal cancer. Upon receiving continuous doses of the drug, a group of liver cells started to generate a physical barrier which stopped cancer cells from being able to invade the organ. The treatment also promoted long-term immune responses that targeted diseased cells while being safe for healthy tissues. If confirmed in clinical trials, these results suggest that colorectal patients undergoing tumor removal surgery may benefit from also receiving interferon-alpha through continuous delivery.


Asunto(s)
Neoplasias Colorrectales , Interferón-alfa , Animales , Ratones , Células Endoteliales/patología , Linfocitos T CD8-positivos , Hígado , Hepatocitos , Neoplasias Colorrectales/patología
11.
J Cell Physiol ; 215(3): 794-802, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18314878

RESUMEN

When expressed in the absence of light chains, secretory Ig-micro chains (micro(s)) undergo endoplasmic reticulum associated degradation (ERAD). This process involves the recognition of terminally misfolded or unassembled molecules, their retro-translocation across the ER membrane and ubiquitination for degradation by cytosolic proteasomes. The molecular components of the ERAD pathway and their coordination remain largely unknown. Here we employed co-immunoprecipitation, silencing or over-expression assays to show that SEL1L and HRD1 are involved in the degradation of unassembled Ig-micro(s), but have minor effects on another substrate, TCR-alpha. SEL1L and HRD1 localize in the early secretory apparatus and are induced by ER stress and during B cell differentiation, concomitantly with the onset of massive IgM secretion. These findings reveal a role for SEL1L and HRD1 in IgM quality control.


Asunto(s)
Inmunoglobulina M/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Regulación de la Expresión Génica , Humanos , Inmunoprecipitación , Unión Proteica , Transporte de Proteínas , Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Ubiquitina-Proteína Ligasas/genética
12.
Sci Rep ; 7: 41815, 2017 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-28157181

RESUMEN

The polymeric structure of secretory IgM allows efficient antigen binding and complement fixation. The available structural models place the N-glycans bound to asparagines 402 and 563 of Ig-µ chains within a densely packed core of native IgM. These glycans are found in the high mannose state also in secreted IgM, suggesting that polymerization hinders them to Golgi processing enzymes. Their absence alters polymerization. Here we investigate their role following the fate of aggregation-prone mutant µ chains lacking the Cµ1 domain (µ∆). Our data reveal that µ∆ lacking 563 glycans (µ∆5) form larger intracellular aggregates than µ∆ and are not secreted. Like µ∆, they sequester ERGIC-53, a lectin previously shown to promote polymerization. In contrast, µ∆ lacking 402 glycans (µ∆4) remain detergent soluble and accumulate in the ER, as does a double mutant devoid of both (µ∆4-5). These results suggest that the two C-terminal Ig-µ glycans shape the polymerization-dependent aggregation by engaging lectins and acting as spacers in the alignment of individual IgM subunits in native polymers.


Asunto(s)
Inmunoglobulina M/metabolismo , Cadenas mu de Inmunoglobulina/metabolismo , Polisacáridos/metabolismo , Agregado de Proteínas , Multimerización de Proteína , Vías Secretoras , Línea Celular , Membrana Celular/metabolismo , Humanos , Inmunoglobulina M/química , Inmunoglobulina M/genética , Cadenas mu de Inmunoglobulina/química , Cadenas mu de Inmunoglobulina/genética , Lectinas/metabolismo , Lectinas de Unión a Manosa/metabolismo , Manosidasas/metabolismo , Proteínas de la Membrana/metabolismo , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas
13.
Antioxid Redox Signal ; 8(3-4): 274-82, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16677073

RESUMEN

Disulfide bonds are formed in the endoplasmic reticulum (ER) by sequential interchange reactions: Ero1alpha and Ero1beta transfer oxidative equivalents to protein disulfide isomerase (PDI), which in turn oxidizes cargo proteins. Neither Ero1alpha nor Ero1beta contains known ER localization motif(s), raising the question of how they are retained in this organelle. Here the authors show that, unlike endogenous molecules, overexpressed Ero1alpha and Ero1beta are secreted by HeLa transfectants, suggesting saturation of their normal retention mechanism(s). Co-expression of either PDI or ERp44 prevents Ero1 secretion in a KDEL/RDEL dependent way. Covalent interactions between ERp44 and Ero1 are essential for retention. In contrast, a mutant PDI lacking the four cysteines in the two active sites still inhibits secretion, albeit less efficiently. PDI and ERp44 compete for Ero1 binding. PDI also prevents Ero1 aggregation and dimerization, thus chaperoning its own oxidase. This dynamic retention mechanism of Ero1 may be important for fine-tuning the regulation of ER redox homeostasis and quality control.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/química , Chaperonas Moleculares/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/fisiología , Oxidorreductasas/fisiología , Proteína Disulfuro Isomerasas/química , Sitios de Unión , Unión Competitiva , Western Blotting , Cisteína/química , Vectores Genéticos , Células HeLa , Humanos , Glicoproteínas de Membrana/química , Oxidación-Reducción , Oxidorreductasas/química , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/química , Plásmidos/metabolismo , Unión Proteica , Transfección
14.
Sci Rep ; 5: 12585, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26223695

RESUMEN

Professional secretory cells produce and release abundant proteins. Particularly in case of mutations and/or insufficient chaperoning, these can aggregate and become toxic within or amongst cells. Immunoglobulins (Ig) are no exception. In the extracellular space, certain Ig-L chains form fibrils causing systemic amyloidosis. On the other hand, Ig variants lacking the first constant domain condense in dilated cisternae of the early secretory compartment, called Russell Bodies (RB), frequently observed in plasma cell dyscrasias, autoimmune diseases and chronic infections. RB biogenesis can be recapitulated in lymphoid and non-lymphoid cells by expressing mutant Ig-µ, providing powerful models to investigate the pathophysiology of endoplasmic reticulum storage disorders. Here we analyze the aggregation propensity and the biochemical features of the intra- and extra-cellular Ig deposits in human cells, revealing ß-aggregated features for RB.


Asunto(s)
Cadenas mu de Inmunoglobulina/metabolismo , Células HEK293 , Células HeLa , Humanos , Cadenas mu de Inmunoglobulina/genética , Microscopía Confocal , Mutación , Espectrofotometría Infrarroja
15.
PLoS One ; 9(10): e108496, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25279560

RESUMEN

Precise coordination of protein biogenesis, traffic and homeostasis within the early secretory compartment (ESC) is key for cell physiology. As a consequence, disturbances in these processes underlie many genetic and chronic diseases. Dynamic imaging methods are needed to follow the fate of cargo proteins and their interactions with resident enzymes and folding assistants. Here we applied the Halotag labelling system to study the behavior of proteins with different fates and roles in ESC: a chaperone, an ERAD substrate and an aggregation-prone molecule. Exploiting the Halo property of binding covalently ligands labelled with different fluorochromes, we developed and performed non-radioactive pulse and chase assays to follow sequential waves of proteins in ESC, discriminating between young and old molecules at the single cell level. In this way, we could monitor secretion and degradation of ER proteins in living cells. We can also follow the biogenesis, growth, accumulation and movements of protein aggregates in the ESC. Our data show that protein deposits within ESC grow by sequential apposition of molecules up to a given size, after which novel seeds are detected. The possibility of using ligands with distinct optical and physical properties offers a novel possibility to dynamically follow the fate of proteins in the ESC.


Asunto(s)
Agregado de Proteínas , Proteínas/metabolismo , Vías Secretoras , Línea Celular , Retículo Endoplásmico/metabolismo , Humanos , Imagen Molecular , Transporte de Proteínas , Proteolisis , Análisis de la Célula Individual
16.
Antioxid Redox Signal ; 16(10): 1077-87, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21854214

RESUMEN

AIMS: The endoplasmic reticulum (ER) is involved in many functions, including protein folding, redox homeostasis, and Ca(2+) storage and signaling. To perform these multiple tasks, the ER is composed of distinct, specialized subregions, amongst which mitochondrial-associated ER membranes (MAM) emerge as key signaling hubs. How these multiple functions are integrated with one another in living cells remains unclear. RESULTS: Here we show that Ero1α, a key controller of oxidative folding and ER redox homeostasis, is enriched in MAM and regulates Ca(2+) fluxes. Downregulation of Ero1α by RNA interference inhibits mitochondrial Ca(2+) fluxes and modifies the activity of mitochondrial Ca(2+) uniporters. The overexpression of redox active Ero1α increases passive Ca(2+) efflux from the ER, lowering [Ca(2+)](ER) and mitochondrial Ca(2+) fluxes in response to IP3 agonists. INNOVATION: The unexpected observation that Ca(2+) fluxes are affected by either increasing or decreasing the levels of Ero1α reveals a pivotal role for this oxidase in the early secretory compartment and implies a strict control of its amounts. CONCLUSIONS: Taken together, our results indicate that the levels, subcellular localization, and activity of Ero1α coordinately regulate Ca(2+) and redox homeostasis and signaling in the early secretory compartment.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Glicoproteínas de Membrana/metabolismo , Mitocondrias/metabolismo , Oxidorreductasas/metabolismo , Línea Celular , Homeostasis , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/metabolismo , Oxidación-Reducción , Oxidorreductasas/genética , Unión Proteica , Transducción de Señal
17.
Mol Immunol ; 47(6): 1356-65, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20044139

RESUMEN

Upon antigen stimulation, B lymphocytes differentiate into antibody secreting cells (ASC), most of which undergo apoptosis after a few days of intense Ig production. Differentiation entails expansion of the endoplasmic reticulum (ER) and requires XBP1 but not other elements of the unfolded protein response, like PERK. Moreover, normal and malignant ASC are exquisitely sensitive to proteasome inhibitors, but the underlying mechanisms are poorly understood. Here we analyze the role of C/EBP homologous protein (CHOP), a transcription factor mediating apoptosis in many cell types that experience high levels of ER stress. CHOP is transiently induced early upon B cell stimulation: covalent IgM aggregates form more readily and IgM secretion is slower in chop(-/-) cells. Despite these subtle changes, ASC differentiation and lifespan are normal in chop(-/-) mice. Unlike fibroblasts and other cell types, chop(-/-) ASC are equally or slightly more sensitive to proteasome inhibitors and ER stressors, implying tissue-specific roles for CHOP in differentiation and stress.


Asunto(s)
Apoptosis , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/metabolismo , Respuesta de Proteína Desplegada , Animales , Diferenciación Celular , Inmunoglobulina M/metabolismo , Ratones , Estrés Fisiológico , Factor de Transcripción CHOP/deficiencia
18.
J Cell Sci ; 119(Pt 12): 2532-41, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16735443

RESUMEN

Owing to the impossibility of reaching the Golgi for secretion or the cytosol for degradation, mutant Ig-mu chains that lack the first constant domain (muDeltaCH1) accumulate as detergent-insoluble aggregates in dilated endoplasmic reticulum cisternae, called Russell bodies. The presence of similar structures hallmarks many ER storage diseases, but their pathogenic role(s) remain obscure. Exploiting inducible cellular systems, we show here that Russell bodies form when the synthesis of muDeltaCH1 exceeds the degradation capacity. Condensation occurs in different sub-cellular locations, depending on the interacting molecules present in the host cell: if Ig light chains are co-expressed, detergent-insoluble muDeltaCH1-light chain oligomers accumulate in large ribosome-coated structures (rough Russell bodies). In absence of light chains, instead, aggregation occurs in smooth tubular vesicles and is controlled by N-glycan-dependent interactions with ER-Golgi intermediate compartment 53 (ERGIC-53). In cells containing smooth Russell bodies, ERGIC-53 co-localizes with muDeltaCH1 aggregates in a Ca2+ -dependent fashion. Our findings identify a novel ERGIC-53 substrate, and indicate that interactions with light chains or ERGIC-53 seed muDeltaCH1 condensation in different stations of the early secretory pathway.


Asunto(s)
Retículo Endoplásmico/metabolismo , Cadenas mu de Inmunoglobulina/metabolismo , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/metabolismo , Errores Innatos del Metabolismo/metabolismo , Alcaloides/farmacología , Sitios de Unión , Calcio/metabolismo , Línea Celular , Retículo Endoplásmico/patología , Células HeLa , Humanos , Cadenas mu de Inmunoglobulina/biosíntesis , Cadenas mu de Inmunoglobulina/efectos de los fármacos , Manosa/antagonistas & inhibidores , Manosa/metabolismo , Lectinas de Unión a Manosa/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Errores Innatos del Metabolismo/patología , Pliegue de Proteína
19.
Biol Chem ; 384(9): 1273-7, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14515988

RESUMEN

B lymphocytes are small cells that express antigen receptors and secrete little if any IgM. Upon encounter with antigen, they differentiate into short-lived plasma cells, which secrete large amounts of polymeric IgM. Plasma cell differentiation entails a massive development of the endoplasmic reticulum to sustain high levels of Ig production. Recent findings suggest a role for the unfolded protein response in orchestrating the architectural and functional changes during terminal plasma cell differentiation.


Asunto(s)
Retículo Endoplásmico/fisiología , Células Plasmáticas/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Retículo Endoplásmico/química , Retículo Endoplásmico/inmunología , Humanos , Células Plasmáticas/inmunología , Pliegue de Proteína
20.
J Biol Chem ; 279(31): 32667-73, 2004 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-15161913

RESUMEN

Many proteins of the secretory pathway contain disulfide bonds that are essential for structure and function. In the endoplasmic reticulum (ER), Ero1 alpha and Ero1 beta oxidize protein disulfide isomerase (PDI), which in turn transfers oxidative equivalents to newly synthesized cargo proteins. However, oxidation must be limited, as some reduced PDI is necessary for disulfide isomerization and ER-associated degradation. Here we show that in semipermeable cells, PDI is more oxidized, disulfide bonds are formed faster, and high molecular mass covalent protein aggregates accumulate in the absence of cytosol. Addition of reduced glutathione (GSH) reduces PDI and restores normal disulfide formation rates. A higher GSH concentration is needed to balance oxidative folding in semipermeable cells overexpressing Ero1 alpha, indicating that cytosolic GSH and lumenal Ero1 alpha play antagonistic roles in controlling the ER redox. Moreover, the overexpression of Ero1 alpha significantly increases the GSH content in HeLa cells. Our data demonstrate tight connections between ER and cytosol to guarantee redox exchange across compartments: a reducing cytosol is important to ensure disulfide isomerization in secretory proteins.


Asunto(s)
Retículo Endoplásmico/metabolismo , Glutatión/metabolismo , Glicoproteínas/metabolismo , Glicoproteínas de Membrana/fisiología , Oxígeno/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Digitonina/farmacología , Disulfuros/metabolismo , Electroforesis en Gel de Poliacrilamida , Vectores Genéticos , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Microscopía Fluorescente , Oxidación-Reducción , Oxidorreductasas , Plásmidos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Pliegue de Proteína , Isoformas de Proteínas , Temperatura , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA