Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell Struct Funct ; 48(2): 123-133, 2023 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-37380437

RESUMO

When medaka fish (Oryzias latipes) larvae are grown in the absence of exogenous nutrition, the liver becomes dark and positive to Oil Red O staining from 7 days post-hatch (dph). We determined the mechanism of this starvation-induced development of fatty liver by proteomic analysis using livers obtained from larvae grown in the presence or absence of 2% glucose at 5 dph. Results showed that changes in the expression levels of enzymes involved in glycolysis or the tricarboxylic acid cycle were modest, whereas the expression levels of enzymes involved in amino acid catabolism or ß-oxidation of fatty acids were significantly elevated, suggesting that they become major energy sources under starvation conditions. Expression levels of enzymes for the uptake and ß-oxidation of fatty acids as well as synthesis of triacylglycerol were elevated, whereas those for the synthesis of cholesterol as well as export of cholesterol and triacylglycerol were decreased under starvation conditions, which explains the accumulation of triacylglycerol in the liver. Our results provide the basis for future research to understand how gene malfunction(s) affects the development of fatty liver, which can lead to nonalcoholic steatohepatitis and then to liver cirrhosis.Key words: amino acid catabolism, ß-oxidation, triacylglycerol, cholesterol, export.


Assuntos
Fígado Gorduroso , Oryzias , Animais , Oryzias/metabolismo , Larva/metabolismo , Proteômica , Fígado Gorduroso/veterinária , Ácidos Graxos/metabolismo , Triglicerídeos/metabolismo , Colesterol , Aminoácidos
2.
Cell Struct Funct ; 45(1): 23-31, 2020 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-31875595

RESUMO

Three types of transmembrane protein, IRE1α/IRE1ß, PERK, and ATF6α/ATF6ß, are expressed ubiquitously in vertebrates as transducers of the unfolded protein response (UPR), which maintains the homeostasis of the endoplasmic reticulum. IRE1 is highly conserved from yeast to mammals, and transmits a signal by a unique mechanism, namely splicing of mRNA encoding XBP1, the transcription factor downstream of IRE1 in metazoans. IRE1 contains a ribonuclease domain in its cytoplasmic region which initiates splicing reaction by direct cleavage of XBP1 mRNA at the two stem loop structures. As the UPR is considered to be involved in the development and progression of various diseases, as well as in the survival and growth of tumor cells, UPR inhibitors have been sought. To date, IRE1 inhibitors have been screened using cell-based reporter assays and fluorescent-based in vitro cleavage assays. Here, we used medaka fish to develop an in vivo assay for IRE1α inhibitors. IRE1α, IRE1ß, ATF6α and ATF6ß are ubiquitously expressed in medaka. We found that IRE1α/ATF6α-double knockout is lethal, similarly to IRE1α/IRE1ß- and ATF6α/ATF6ß-double knockout. Therefore, IRE1 inhibitors are expected to confer lethality to ATF6α-knockout medaka but not to wild-type medaka. One compound named K114 was obtained from 1,280 compounds using this phenotypic screening. K114 inhibited ER stress-induced splicing of XBP1 mRNA as well as reporter luciferase expression in HCT116 cells derived from human colorectal carcinoma, and inhibited ribonuclease activity of human IRE1α in vitro. Thus, this phenotypic assay can be used as a quick test for the efficacy of IRE1α inhibitors in vivo.Key words: endoplasmic reticulum, inhibitor screening, mRNA splicing, phenotypic assay, unfolded protein response.


Assuntos
Endonucleases/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Animais , Endonucleases/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Oryzias , Proteínas Serina-Treonina Quinases/genética , Fatores de Tempo
3.
Cell Struct Funct ; 45(1): 9-21, 2020 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-31852864

RESUMO

ATF6α is an endoplasmic reticulum (ER)-embedded transcription factor which is rapidly activated by ER stress, and a major regulator of ER chaperone levels in vertebrates. We previously suggested that ATF6α occurs as a monomer, dimer and oligomer in the unstressed ER of Chinese hamster ovary cells due to the presence of two evolutionarily conserved cysteine residues in its luminal region (C467 and C618), and showed that ATF6α is reduced upon ER stress, such that only reduced monomer ATF6α is translocated to the Golgi apparatus for activation by proteolysis. However, mutagenesis analysis (C467A and C618A) revealed that the C618A mutant behaves in an unexpected manner (monomer and oligomer) during non-reducing SDS-PAGE, for reasons which remained unclear. Here, we used human colorectal carcinoma-derived HCT116 cells deficient in ATF6α and its relevant ATF6ß, and found that ATF6α dimer and oligomer are both dimers, which we designated C618-dimer and C467-dimer, respectively. We demonstrated that C467-dimer (previously considered an oligomer) behaved bigger than C618-dimer (previously considered a dimer) during non-reducing SDS-PAGE, based on their disulfide-bonded structures. Furthermore, ATF6α monomer physically associates with another ATF6α monomer in the absence of disulfide bonding, which renders two C467 residues in close proximity so that formation of C467-dimer is much easier than that of C618-dimer. In contrast, C618-dimer is more easily reduced upon ER stress. Thus, our analysis revealed that all forms of ATF6α, namely monomer, C618-dimer and C467-dimer, are activated by single reduction of a disulfide bond in response to ER stress, ensuring the rapidity of ATF6α activation.Key words: disulfide-bonded structure, endoplasmic reticulum, membrane-bound transcription factor, non-reducing SDS-PAGE, unfolded protein response.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Dissulfetos/metabolismo , Retículo Endoplasmático/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Fator 6 Ativador da Transcrição/genética , Animais , Cricetinae , Cricetulus/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Regulação da Expressão Gênica/fisiologia , Complexo de Golgi/metabolismo , Humanos , Chaperonas Moleculares/metabolismo
4.
Mol Cell Proteomics ; 15(11): 3424-3434, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27601598

RESUMO

Dystroglycanopathy is a major class of congenital muscular dystrophy caused by a deficiency of functional glycans on α-dystroglycan (αDG) with laminin-binding activity. Recent advances have led to identification of several causative gene products of dystroglycanopathy and characterization of their in vitro enzymatic activities. However, the in vivo functional roles remain equivocal for enzymes such as ISPD, FKTN, FKRP, and TMEM5 that are supposed to be involved in post-phosphoryl modifications linking the GalNAc-ß3-GlcNAc-ß4-Man-6-phosphate core and the outer laminin-binding glycans. Herein, by direct nano-LC-MS2/MS3 analysis of tryptic glycopeptides derived from a truncated recombinant αDG expressed in the wild-type and a panel of mutated cells deficient in one of these enzymes, we sought to define the full extent of variable modifications on this phosphorylated core O-glycan at the functional Thr317/Thr319 sites. We showed that the most abundant glycoforms carried a phosphorylated core at each of the two sites, with and without a single ribitol phosphate (RboP) extending from terminal HexNAc. At much lower signal intensity, a novel substituent tentatively assigned as glycerol phosphate (GroP) was additionally detected. As expected, tandem RboP extended with a GlcA-Xyl unit was only identified in wild type, whereas knocking out of either ISPD or FKTN prevented formation of RboP. In the absence of FKRP, glycoforms with single but not tandem RboP accumulated, consistent with the suggested role of this enzyme in transferring the second RboP. Intriguingly, the single GroP modification also required functional FKTN whereas absence of TMEM5 significantly hindered only the addition of RboP. Our findings thus revealed additional levels of complexity associated with the core structures, suggesting functional interplay among these enzymes through their interactions. The simplified analytical workflow developed here should facilitate rapid mapping across a wider range of cell types to gain better insights into its physiological relevance.


Assuntos
Distroglicanas/química , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , Síndrome de Walker-Warburg/genética , Técnicas de Inativação de Genes , Predisposição Genética para Doença , Células HCT116 , Células HEK293 , Humanos , Pentosiltransferases , Fosforilação , Proteínas/genética
5.
Cell Struct Funct ; 42(2): 81-94, 2017 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-28552883

RESUMO

Accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) activates the unfolded protein response (UPR). The ATF6 pathway is one of the three major pathways in vertebrates. Although ATF6, a transmembrane-type glycoprotein in the ER, functions as a UPR sensor/transducer, it is an unstable protein with a half-life of approximately 2 h and is constitutively subjected to the ER-associated degradation system with the location of the misfolded part in the ER lumen (ERAD-L). ERAD-L substrates are delivered to the cytosol through the retrotranslocon, which likely contains HRD1 (E3), gp78 (E3), SEL1L (a partner of HRD1), Derlin1/2/3 and Herp1/2. We previously showed that ATF6 represents a novel transmembrane-type ERAD-L substrate requiring both EDEM1/2/3-mediated mannose trimming and SEL1L. Here, by constructing and analyzing chicken DT40 cells deficient in various components of the retrotranslocon, we show that degradation of ATF6 requires Derlin2 or Derlin3 and that Derlin2 and Derlin3 are redundant for ERAD-L of ATF6. We further show that degradation of ATF6 requires Herp1 or Herp2 and that Herp1 and Herp2 are redundant for ERAD-L of ATF6. Furthermore, by investigating five more ERAD-L substrates, we show that SEL1L-dependent substrates require Derlin2/3 and Herp1/2 regardless of their soluble or transmembrane nature. Our results suggest that ERAD-L substrates take several routes to the cytosol. The HRD1-engaged route 1 requires SEL1L, Derlin2 or Derlin3, and Herp1 or Herp2, whereas the HRD1-engaged route 2 does not require them functionally. It remains to be determined whether the latter requires Derlin1 and whether these two routes are compositionally distinct.Key words: endoplasmic reticulum, proteasome, protein degradation, protein misfolding, ubiquitin.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Membrana/metabolismo , Proteólise , Proteínas Repressoras/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Células Cultivadas , Galinhas , Resposta a Proteínas não Dobradas
6.
J Biol Chem ; 288(44): 31517-27, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24043630

RESUMO

Proteins misfolded in the endoplasmic reticulum (ER) are cleared by the ubiquitin-dependent proteasome system in the cytosol, a series of events collectively termed ER-associated degradation (ERAD). It was previously shown that SEL1L, a partner protein of the E3 ubiquitin ligase HRD1, is required for degradation of misfolded luminal proteins (ERAD-Ls substrates) but not misfolded transmembrane proteins (ERAD-Lm substrates) in both mammalian and chicken DT40 cells. Here, we analyzed ATF6, a type II transmembrane glycoprotein that serves as a sensor/transducer of the unfolded protein response, as a potential ERAD-Lm substrate in DT40 cells. Unexpectedly, degradation of endogenous ATF6 and exogenously expressed chicken and human ATF6 by the proteasome required SEL1L. Deletion analysis revealed that the luminal region of ATF6 is a determinant for SEL1L-dependent degradation. Chimeric analysis showed that the luminal region of ATF6 confers SEL1L dependence on type I transmembrane protein as well. In contrast, degradation of other known type I ERAD-Lm substrates (BACE457, T-cell receptor-α, CD3-δ, and CD147) did not require SEL1L. Thus, ATF6 represents a novel type of ERAD-Lm substrate requiring SEL1L for degradation despite its transmembrane nature. In addition, endogenous ATF6 was markedly stabilized in wild-type cells treated with kifunensine, an inhibitor of α1,2-mannosidase in the ER, indicating that degradation of ATF6 requires proper mannose trimming. Our further analyses revealed that the five ERAD-Lm substrates examined are classified into three subgroups based on their dependence on mannose trimming and SEL1L. Thus, ERAD-Lm substrates are degraded through much more diversified mechanisms in higher eukaryotes than previously thought.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Degradação Associada com o Retículo Endoplasmático/fisiologia , Manose/metabolismo , Manosidases/metabolismo , Proteínas/metabolismo , Fator 6 Ativador da Transcrição/genética , Alcaloides/farmacologia , Animais , Antidepressivos/farmacologia , Linhagem Celular , Galinhas , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Manose/genética , Manosidases/antagonistas & inibidores , Manosidases/genética , Fenelzina/farmacologia , Estabilidade Proteica/efeitos dos fármacos , Proteínas/genética , Proteólise/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/fisiologia
7.
Mol Biol Cell ; 34(3): ar20, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36696173

RESUMO

To survive poor nutritional conditions, tumor cells activate the unfolded protein response, which is composed of the IRE1, PERK, and ATF6 arms, to maintain the homeostasis of the endoplasmic reticulum, where secretory and transmembrane proteins destined for the secretory pathway gain their correct three-dimensional structure. The requirement of the IRE1 and PERK arms for tumor growth in nude mice is established. Here we investigated the requirement for the ATF6 arm, which consists of ubiquitously expressed ATF6α and ATF6ß, by constructing ATF6α-knockout (KO), ATF6ß-KO, and ATF6α/ß-double KO (DKO) in HCT116 cells derived from human colorectal carcinoma. Results showed that these KO cells grew similarly to wild-type (WT) cells in nude mice, contrary to expectations from our analysis of ATF6α-KO, ATF6ß-KO, and ATF6α/ß-DKO mice. We then found that the loss of ATF6α in HCT116 cells resulted in sustained activation of the IRE1 and PERK arms in marked contrast to mouse embryonic fibroblasts, in which the loss of ATF6α is compensated for by ATF6ß. Although IRE1-KO in HCT116 cells unexpectedly did not affect tumor growth in nude mice, IRE1-KO HCT116 cells with ATF6α knockdown grew significantly more slowly than WT or IRE1-KO HCT116 cells. These results have unraveled the situation-dependent differential compensation strategies of ATF6α.


Assuntos
Carcinoma , Fibroblastos , Animais , Humanos , Camundongos , Fator 6 Ativador da Transcrição/metabolismo , Linhagem Celular , Estresse do Retículo Endoplasmático , Fibroblastos/metabolismo , Camundongos Nus , Proteínas Serina-Treonina Quinases/metabolismo , Resposta a Proteínas não Dobradas
8.
Elife ; 112022 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-36444643

RESUMO

A causal relationship between endoplasmic reticulum (ER) stress and the development of neurodegenerative diseases remains controversial. Here, we focused on Seipinopathy, a dominant motor neuron disease, based on the finding that its causal gene product, Seipin, is a protein that spans the ER membrane twice. Gain-of-function mutations of Seipin produce non-glycosylated Seipin (ngSeipin), which was previously shown to induce ER stress and apoptosis at both cell and mouse levels albeit with no clarified mechanism. We found that aggregation-prone ngSeipin dominantly inactivated SERCA2b, the major calcium pump in the ER, and decreased the calcium concentration in the ER, leading to ER stress and apoptosis in human colorectal carcinoma-derived cells (HCT116). This inactivation required oligomerization of ngSeipin and direct interaction of the C-terminus of ngSeipin with SERCA2b, and was observed in Seipin-deficient neuroblastoma (SH-SY5Y) cells expressing ngSeipin at an endogenous protein level. Our results thus provide a new direction to the controversy noted above.


Assuntos
Doença dos Neurônios Motores , Neuroblastoma , Humanos , Animais , Camundongos , Cálcio , Neuroblastoma/genética , Apoptose , Mutação
9.
Cell Struct Funct ; 36(2): 187-95, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21857145

RESUMO

Proteins misfolded in the endoplasmic reticulum (ER) are degraded in the cytosol by a ubiquitin-dependent proteasome system, a process collectively termed ER-associated degradation (ERAD). Unraveling the molecular mechanisms of mammalian ERAD progresses more slowly than that of yeast ERAD due to the laborious procedures required for gene targeting and the redundancy of components. Here, we utilized the chicken B lymphocyte-derived DT40 cell line, which exhibits an extremely high homologous recombination frequency, to analyze ERAD mechanisms in higher eukaryotes. We disrupted the SEL1L gene, which encodes the sole homologue of yeast Hrd3p in both chickens and mammals; Hrd3p is a binding partner of yeast Hrd1p, an E3 ubiquitin ligase. SEL1L-knockout cells grew only slightly more slowly than the wild-type cells. Pulse chase experiments revealed that chicken SEL1L was required for ERAD of misfolded luminal proteins such as glycosylated NHK and unglycosylated NHK-QQQ but dispensable for that of misfolded transmembrane proteins such as NHK(BACE) and CD3-δ, as in mammals. The defect of SEL1L-knockout cells in NHK degradation was restored by introduction of not only chicken SEL1L but also mouse and human SEL1L. Deletion analysis showed the importance of Sel1-like tetratricopeptide repeats but not the fibronectin II domain in the function of SEL1L. Thus, our reverse genetic approach using the chicken DT40 cell line will provide highly useful information regarding ERAD mechanisms in higher eukaryotes which express ERAD components redundantly.


Assuntos
Retículo Endoplasmático/metabolismo , Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Galinhas , Retículo Endoplasmático/enzimologia , Técnicas de Inativação de Genes , Glicosilação , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Complexo de Endopeptidases do Proteassoma/metabolismo , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas/química , Proteínas/genética , Alinhamento de Sequência , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/metabolismo , alfa 1-Antitripsina/genética , alfa 1-Antitripsina/metabolismo
11.
Biochim Biophys Acta Gen Subj ; 1865(3): 129812, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33316349

RESUMO

BACKGROUND: The quality of proteins destined for the secretory pathway is ensured by two distinct mechanisms in the endoplasmic reticulum (ER): productive folding of newly synthesized proteins, which is assisted by ER-localized molecular chaperones and in most cases also by disulfide bond formation and transfer of an oligosaccharide unit; and ER-associated degradation (ERAD), in which proteins unfolded or misfolded in the ER are recognized and processed for delivery to the ER membrane complex, retrotranslocated through the complex with simultaneous ubiquitination, extracted by AAA-ATPase to the cytosol, and finally degraded by the proteasome. SCOPE OF REVIEW: We describe the mechanisms of productive folding and ERAD, with particular attention to glycoproteins versus non-glycoproteins, and to yeast versus mammalian systems. MAJOR CONCLUSION: Molecular mechanisms of the productive folding of glycoproteins and non-glycoproteins mediated by molecular chaperones and protein disulfide isomerases are well conserved from yeast to mammals. Additionally, mammals have gained an oligosaccharide structure-dependent folding cycle for glycoproteins. The molecular mechanisms of ERAD are also well conserved from yeast to mammals, but redundant expression of yeast orthologues in mammals has been encountered, particularly for components involved in recognition and processing of glycoproteins and components of the ER membrane complex involved in retrotranslocation and simultaneous ubiquitination of glycoproteins and non-glycoproteins. This may reflect an evolutionary consequence of increasing quantity or quality needs toward mammals. GENERAL SIGNIFICANCE: The introduction of innovative genome editing technology into analysis of the mechanisms of mammalian ERAD, as exemplified here, will provide new insights into the pathogenesis of various diseases.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Glicoproteínas/metabolismo , Chaperonas Moleculares/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Retículo Endoplasmático/genética , Edição de Genes/métodos , Glicoproteínas/genética , Glicosilação , Chaperonas Moleculares/genética , Oligossacarídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Biossíntese de Proteínas , Isomerases de Dissulfetos de Proteínas/genética , Dobramento de Proteína , Transporte Proteico , Proteólise , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ubiquitinação
12.
Elife ; 102021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34698634

RESUMO

Sequential mannose trimming of N-glycan, from M9 to M8B and then to oligosaccharides exposing the α1,6-linked mannosyl residue (M7A, M6, and M5), facilitates endoplasmic reticulum-associated degradation of misfolded glycoproteins (gpERAD). We previously showed that EDEM2 stably disulfide-bonded to the thioredoxin domain-containing protein TXNDC11 is responsible for the first step (George et al., 2020). Here, we show that EDEM3 and EDEM1 are responsible for the second step. Incubation of pyridylamine-labeled M8B with purified EDEM3 alone produced M7 (M7A and M7C), M6, and M5. EDEM1 showed a similar tendency, although much lower amounts of M6 and M5 were produced. Thus, EDEM3 is a major α1,2-mannosidase for the second step from M8B. Both EDEM3 and EDEM1 trimmed M8B from a glycoprotein efficiently. Our confirmation of the Golgi localization of MAN1B indicates that no other α1,2-mannosidase is required for gpERAD. Accordingly, we have established the entire route of oligosaccharide processing and the enzymes responsible.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Degradação Associada com o Retículo Endoplasmático/genética , Glicoproteínas/metabolismo , Proteínas de Membrana/genética , Oligossacarídeos/metabolismo , alfa-Manosidase/genética , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular , Humanos , Proteínas de Membrana/metabolismo , alfa-Manosidase/metabolismo
13.
Elife ; 92020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33198886

RESUMO

Second-generation antipsychotics are widely used to medicate patients with schizophrenia, but may cause metabolic side effects such as diabetes, which has been considered to result from obesity-associated insulin resistance. Olanzapine is particularly well known for this effect. However, clinical studies have suggested that olanzapine-induced hyperglycemia in certain patients cannot be explained by such a generalized mechanism. Here, we focused on the effects of olanzapine on insulin biosynthesis and secretion by mouse insulinoma MIN6 cells. Olanzapine reduced maturation of proinsulin, and thereby inhibited secretion of insulin; and specifically shifted the primary localization of proinsulin from insulin granules to the endoplasmic reticulum. This was due to olanzapine's impairment of proper disulfide bond formation in proinsulin, although direct targets of olanzapine remain undetermined. Olanzapine-induced proinsulin misfolding and subsequent decrease also occurred at the mouse level. This mechanism of olanzapine-induced ß-cell dysfunction should be considered, together with weight gain, when patients are administered olanzapine.


Assuntos
Diabetes Mellitus/induzido quimicamente , Retículo Endoplasmático/metabolismo , Olanzapina/toxicidade , Proinsulina/metabolismo , Dobramento de Proteína/efeitos dos fármacos , Animais , Antipsicóticos/toxicidade , Linhagem Celular Tumoral , Diabetes Mellitus/metabolismo , Insulinoma , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Risperidona/toxicidade
14.
Nat Commun ; 11(1): 1368, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32170195

RESUMO

MCFD2 and ERGIC-53, which are the products of causative genes of combined factor V and factor VIII deficiency, form a cargo receptor complex responsible for intracellular transport of these coagulation factors in the early secretory pathway. In this study, using an NMR technique, we successfully identified an MCFD2-binding segment from factor VIII composed of a 10 amino acid sequence that enhances its secretion. This prompted us to examine possible effects of attaching this sequence to recombinant glycoproteins on their secretion. We found that the secretion level of recombinant erythropoietin was significantly increased simply by tagging it with the passport sequence. Our findings not only provide molecular basis for the intracellular trafficking of coagulation factors and their genetic deficiency but also offer a potentially useful tool for increasing the production yields of recombinant glycoproteins of biopharmaceutical interest.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas/metabolismo , Proteínas de Membrana/metabolismo , Polissacarídeos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Retículo Endoplasmático/fisiologia , Eritropoetina/metabolismo , Fator V , Fator VIII/metabolismo , Glicoproteínas/genética , Complexo de Golgi/fisiologia , Humanos , Lectinas de Ligação a Manose/metabolismo , Transporte Proteico , Via Secretória
15.
Elife ; 92020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-32065582

RESUMO

Sequential mannose trimming of N-glycan (Man9GlcNAc2 -> Man8GlcNAc2 -> Man7GlcNAc2) facilitates endoplasmic reticulum-associated degradation of misfolded glycoproteins (gpERAD). Our gene knockout experiments in human HCT116 cells have revealed that EDEM2 is required for the first step. However, it was previously shown that purified EDEM2 exhibited no α1,2-mannosidase activity toward Man9GlcNAc2 in vitro. Here, we found that EDEM2 was stably disulfide-bonded to TXNDC11, an endoplasmic reticulum protein containing five thioredoxin (Trx)-like domains. C558 present outside of the mannosidase homology domain of EDEM2 was linked to C692 in Trx5, which solely contains the CXXC motif in TXNDC11. This covalent bonding was essential for mannose trimming and subsequent gpERAD in HCT116 cells. Furthermore, EDEM2-TXNDC11 complex purified from transfected HCT116 cells converted Man9GlcNAc2 to Man8GlcNAc2(isomerB) in vitro. Our results establish the role of EDEM2 as an initiator of gpERAD, and represent the first clear demonstration of in vitro mannosidase activity of EDEM family proteins.


Assuntos
Proteínas de Transporte/metabolismo , Degradação Associada com o Retículo Endoplasmático , Glicoproteínas/metabolismo , Manose/metabolismo , alfa-Manosidase/metabolismo , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Catálise , Edição de Genes , Técnicas de Silenciamento de Genes , Células HCT116 , Humanos , Manosidases/metabolismo , Reação em Cadeia da Polimerase
16.
J Cell Biol ; 216(6): 1761-1774, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28500182

RESUMO

The unfolded protein response (UPR) handles unfolded/misfolded proteins accumulated in the endoplasmic reticulum (ER). However, it is unclear how vertebrates correctly use the total of ten UPR transducers. We have found that ER stress occurs physiologically during early embryonic development in medaka fish and that the smooth alignment of notochord cells requires ATF6 as a UPR transducer, which induces ER chaperones for folding of type VIII (short-chain) collagen. After secretion of hedgehog for tissue patterning, notochord cells differentiate into sheath cells, which synthesize type II collagen. In this study, we show that this vacuolization step requires both ATF6 and BBF2H7 as UPR transducers and that BBF2H7 regulates a complete set of genes (Sec23/24/13/31, Tango1, Sedlin, and KLHL12) essential for the enlargement of COPII vesicles to accommodate long-chain collagen for export, leading to the formation of the perinotochordal basement membrane. Thus, the most appropriate UPR transducer is activated to cope with the differing physiological ER stresses of different content types depending on developmental stage.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Colágeno Tipo II/metabolismo , Proteínas de Peixes/metabolismo , Notocorda/metabolismo , Oryzias/metabolismo , Resposta a Proteínas não Dobradas , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Animais , Animais Geneticamente Modificados , Membrana Basal/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Embrião não Mamífero/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Células HCT116 , Humanos , Oryzias/embriologia , Oryzias/genética , Fenótipo , Transporte Proteico , Fatores de Tempo , Transcrição Gênica , Transfecção , Vacúolos/metabolismo
17.
J Cell Biol ; 211(4): 775-84, 2015 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-26572623

RESUMO

Glycoproteins and non-glycoproteins possessing unfolded/misfolded parts in their luminal regions are cleared from the endoplasmic reticulum (ER) by ER-associated degradation (ERAD)-L with distinct mechanisms. Two-step mannose trimming from Man9GlcNAc2 is crucial in the ERAD-L of glycoproteins. We recently showed that this process is initiated by EDEM2 and completed by EDEM3/EDEM1. Here, we constructed chicken and human cells simultaneously deficient in EDEM1/2/3 and analyzed the fates of four ERAD-L substrates containing three potential N-glycosylation sites. We found that native but unstable or somewhat unfolded glycoproteins, such as ATF6α, ATF6α(C), CD3-δ-ΔTM, and EMC1, were stabilized in EDEM1/2/3 triple knockout cells. In marked contrast, degradation of severely misfolded glycoproteins, such as null Hong Kong (NHK) and deletion or insertion mutants of ATF6α(C), CD3-δ-ΔTM, and EMC1, was delayed only at early chase periods, but they were eventually degraded as in wild-type cells. Thus, higher eukaryotes are able to extract severely misfolded glycoproteins from glycoprotein ERAD and target them to the non-glycoprotein ERAD pathway to maintain the homeostasis of the ER.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Glicoproteínas/metabolismo , Fator 6 Ativador da Transcrição/metabolismo , Proteínas de Ligação ao Cálcio/genética , Estresse do Retículo Endoplasmático , Técnicas de Inativação de Genes , Glicoproteínas/genética , Células HCT116 , Humanos , Manosidases/genética , Proteínas de Membrana/genética , Dobramento de Proteína , alfa-Manosidase/genética
18.
J Cell Biol ; 206(3): 347-56, 2014 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-25092655

RESUMO

Glycoproteins misfolded in the endoplasmic reticulum (ER) are subjected to ER-associated glycoprotein degradation (gpERAD) in which Htm1-mediated mannose trimming from the oligosaccharide Man8GlcNAc2 to Man7GlcNAc2 is the rate-limiting step in yeast. In contrast, the roles of the three Htm1 homologues (EDEM1/2/3) in mammalian gpERAD have remained elusive, with a key controversy being whether EDEMs function as mannosidases or as lectins. We therefore conducted transcription activator-like effector nuclease-mediated gene knockout analysis in human cell line and found that all endogenous EDEMs possess mannosidase activity. Mannose trimming from Man8GlcNAc2 to Man7GlcNAc2 is performed mainly by EDEM3 and to a lesser extent by EDEM1. Most surprisingly, the upstream mannose trimming from Man9GlcNAc2 to Man8GlcNAc2 is conducted mainly by EDEM2, which was previously considered to lack enzymatic activity. Based on the presence of two rate-limiting steps in mammalian gpERAD, we propose that mammalian cells double check gpERAD substrates before destruction by evolving EDEM2, a novel-type Htm1 homologue that catalyzes the first mannose trimming step from Man9GlcNAc2.


Assuntos
Proteínas Aviárias/fisiologia , Degradação Associada com o Retículo Endoplasmático , Glicoproteínas/fisiologia , Manose/metabolismo , alfa-Manosidase/fisiologia , Sequência de Aminoácidos , Animais , Galinhas , Sequência Conservada , Glicosilação , Células HCT116 , Humanos , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA